551
|
Robust Enhancement of Neural Differentiation from Human ES and iPS Cells Regardless of their Innate Difference in Differentiation Propensity. Stem Cell Rev Rep 2010; 6:270-81. [DOI: 10.1007/s12015-010-9138-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
552
|
Pekkanen-Mattila M, Pelto-Huikko M, Kujala V, Suuronen R, Skottman H, Aalto-Setälä K, Kerkelä E. Spatial and temporal expression pattern of germ layer markers during human embryonic stem cell differentiation in embryoid bodies. Histochem Cell Biol 2010; 133:595-606. [PMID: 20369364 DOI: 10.1007/s00418-010-0689-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
Human embryonic stem cell (hESC) differentiation in embryoid bodies (EBs) provides a valuable tool to study the interplay of different germ layers and their influence on cell differentiation. The gene expression of the developing EBs has been shown in many studies, but the protein expression and the spatial composition of different germ layers in human EBs have not been systematically studied. The aim of the present work was to study the temporal and spatial organisation of germ layers based on the expression of mesoderm (Brachyury T), endoderm (AFP) and ectoderm (SOX1) markers during the early stages of differentiation in eight hESC lines. Tissue multi-array technology was applied to study the protein expression of a large number of EBs. According to our results, EB formation and the organisation of germ layers occurred in a similar manner in all the lines. During 12 days of differentiation, all the germ layer markers were present, but no obvious distinct trajectories were formed. However, older EBs were highly organised in structure. Pluripotency marker OCT3/4 expression persisted unexpectedly long in the differentiating EBs. Cavity formation was observed in the immunocytological sections, and caspase-3 expression was high, suggesting a role of apoptosis in hESC differentiation and/or EB formation. The expression of Brachyury T was notably low in all the lines, also those with the best cardiac differentiation capacity, while the expression of SOX1 was higher in some lines, suggesting that the neural differentiation propensity may be detectable already in the early stages of EB differentiation.
Collapse
Affiliation(s)
- Mari Pekkanen-Mattila
- Regea Institute for Regenerative Medicine, University of Tampere, Tampere University Hospital, Biokatu 12, 33520, Tampere, Finland
| | | | | | | | | | | | | |
Collapse
|
553
|
Närvä E, Autio R, Rahkonen N, Kong L, Harrison N, Kitsberg D, Borghese L, Itskovitz-Eldor J, Rasool O, Dvorak P, Hovatta O, Otonkoski T, Tuuri T, Cui W, Brüstle O, Baker D, Maltby E, Moore HD, Benvenisty N, Andrews PW, Yli-Harja O, Lahesmaa R. High-resolution DNA analysis of human embryonic stem cell lines reveals culture-induced copy number changes and loss of heterozygosity. Nat Biotechnol 2010; 28:371-7. [PMID: 20351689 DOI: 10.1038/nbt.1615] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 02/16/2010] [Indexed: 11/08/2022]
Abstract
Prolonged culture of human embryonic stem cells (hESCs) can lead to adaptation and the acquisition of chromosomal abnormalities, underscoring the need for rigorous genetic analysis of these cells. Here we report the highest-resolution study of hESCs to date using an Affymetrix SNP 6.0 array containing 906,600 probes for single nucleotide polymorphisms (SNPs) and 946,000 probes for copy number variations (CNVs). Analysis of 17 different hESC lines maintained in different laboratories identified 843 CNVs of 50 kb-3 Mb in size. We identified, on average, 24% of the loss of heterozygosity (LOH) sites and 66% of the CNVs changed in culture between early and late passages of the same lines. Thirty percent of the genes detected within CNV sites had altered expression compared to samples with normal copy number states, of which >44% were functionally linked to cancer. Furthermore, LOH of the q arm of chromosome 16, which has not been observed previously in hESCs, was detected.
Collapse
Affiliation(s)
- Elisa Närvä
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
554
|
|
555
|
|
556
|
Fisher OZ, Khademhosseini A, Langer R, Peppas NA. Bioinspired materials for controlling stem cell fate. Acc Chem Res 2010; 43:419-28. [PMID: 20043634 DOI: 10.1021/ar900226q] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although researchers currently have limited ability to mimic the natural stem cell microenvironment, recent work at the interface of stem biology and biomaterials science has demonstrated that control over stem cell behavior with artificial microenvironments is quite advanced. Embryonic and adult stem cells are potentially useful platforms for tissue regeneration, cell-based therapeutics, and disease-in-a-dish models for drug screening. The major challenge in this field is to reliably control stem cell behavior outside the body. Common biological control schemes often ignore physicochemical parameters that materials scientists and engineers commonly manipulate, such as substrate topography and mechanical and rheological properties. However, with appropriate attention to these parameters, researchers have designed novel synthetic microenvironments to control stem cell behavior in rather unnatural ways. In this Account, we review synthetic microenvironments that aim to overcome the limitations of natural niches rather than to mimic them. A biomimetic stem cell control strategy is often limited by an incomplete understanding of the complex signaling pathways that drive stem cell behavior from early embryogenesis to late adulthood. The stem cell extracellular environment presents a miscellany of competing biological signals that keep the cell in a state of unstable equilibrium. Using synthetic polymers, researchers have designed synthetic microenvironments with an uncluttered array of cell signals, both specific and nonspecific, that are motivated by rather than modeled after biology. These have proven useful in maintaining cell potency, studying asymmetric cell division, and controlling cellular differentiation. We discuss recent research that highlights important biomaterials properties for controlling stem cell behavior, as well as advanced processes for selecting those materials, such as combinatorial and high-throughput screening. Much of this work has utilized micro- and nanoscale fabrication tools for controlling material properties and generating diversity in both two and three dimensions. Because of their ease of synthesis and similarity to biological soft matter, hydrogels have become a biomaterial of choice for generating 3D microenvironments. In presenting these efforts within the framework of synthetic biology, we anticipate that future researchers may exploit synthetic polymers to create microenvironments that control stem cell behavior in clinically relevant ways.
Collapse
Affiliation(s)
| | - Ali Khademhosseini
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology
- Department of Chemical Engineering
| | - Nicholas A. Peppas
- Departments of Chemical Engineering and Biomedical Engineering and Division of Pharmaceutics, The University of Texas at Austin, Austin, Texas 78712
| |
Collapse
|
557
|
Abstract
Induction of pluripotency from somatic cells by exogenous transcription factors is made possible by a variety of epigenetic changes that take place during the reprogramming process. The derivation of fully reprogrammed induced pluripotent stem (iPS) cells is achieved through establishment of embryonic stem cell (ESC)-like epigenetic architecture permitting the reactivation of key endogenous pluripotency-related genes, establishment of appropriate bivalent chromatin domains and DNA hypomethylation of genomic heterochromatic regions. Restructuring of the epigenetic landscape, however, is a very inefficient process and the vast majority of the induced cells fail to complete the reprogramming process. Optimal ESC-like epigenetic reorganization is necessary for all reliable downstream uses of iPS cells, including in vitro modeling of disease and clinical applications. Here, we discuss the key advancements in the understanding of dynamic epigenetic changes taking place over the course of the reprogramming process and how aberrant epigenetic remodeling may impact downstream applications of iPS cell technology.
Collapse
|
558
|
Yechoor V, Chan L. Minireview: beta-cell replacement therapy for diabetes in the 21st century: manipulation of cell fate by directed differentiation. Mol Endocrinol 2010; 24:1501-11. [PMID: 20219891 DOI: 10.1210/me.2009-0311] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic beta-cell failure underlies type 1 diabetes; it also contributes in an essential way to type 2 diabetes. beta-Cell replacement is an important component of any cure for diabetes. The current options of islet and pancreas transplantation are not satisfactory as definitive forms of therapy. Here, we review strategies for induced de novo pancreatic beta-cell formation, which depend on the targeted differentiation of cells into pancreatic beta-cells. With this objective in mind, one can manipulate the fate of three different types of cells: 1) from terminally differentiated cells, e.g. exocrine pancreatic cells, into beta-cells; 2) from multipotent adult stem cells, e.g. hepatic oval cells, into pancreatic islets; and 3) from pluripotent stem cells, e.g. embryonic stem cells and induced pluripotent stem cells, into beta-cells. We will examine the pros and cons of each strategy as well as the hurdles that must be overcome before these approaches to generate new beta-cells will be ready for clinical application.
Collapse
Affiliation(s)
- Vijay Yechoor
- One Baylor Plaza, R614, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
559
|
Jia F, Wilson KD, Sun N, Gupta DM, Huang M, Li Z, Robbins RC, Kay MA, Longaker MT, Wu JC. A nonviral minicircle vector for deriving human iPS cells. Nat Methods 2010; 7:197-9. [PMID: 20139967 PMCID: PMC2892897 DOI: 10.1038/nmeth.1426] [Citation(s) in RCA: 507] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 01/13/2010] [Indexed: 12/31/2022]
Abstract
Owing to the risk of insertional mutagenesis, viral transduction has been increasingly replaced by nonviral methods to generate induced pluripotent stem cells (iPSCs). We report the use of 'minicircle' DNA, a vector type that is free of bacterial DNA and capable of high expression in cells, for this purpose. Here we use a single minicircle vector to generate transgene-free iPSCs from adult human adipose stem cells.
Collapse
Affiliation(s)
- Fangjun Jia
- Departments of Radiology and Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Kitchener D. Wilson
- Departments of Radiology and Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Ning Sun
- Departments of Radiology and Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Deepak M. Gupta
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Mei Huang
- Departments of Radiology and Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Zongjin Li
- Departments of Radiology and Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Robert C. Robbins
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Mark A. Kay
- Departments of Pediatrics and Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Michael T. Longaker
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph C. Wu
- Departments of Radiology and Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
560
|
Seifinejad A, Tabebordbar M, Baharvand H, Boyer LA, Hosseini Salekdeh G. Progress and Promise Towards Safe Induced Pluripotent Stem Cells for Therapy. Stem Cell Rev Rep 2010; 6:297-306. [DOI: 10.1007/s12015-010-9121-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
561
|
Derivation of new human embryonic stem cell lines from preimplantation genetic screening and diagnosis-analyzed embryos. In Vitro Cell Dev Biol Anim 2010; 46:395-402. [DOI: 10.1007/s11626-010-9293-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Accepted: 01/14/2010] [Indexed: 12/30/2022]
|
562
|
Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc Natl Acad Sci U S A 2010. [PMID: 20160098 DOI: 10.1073/pnas.01910012107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
For the promise of human induced pluripotent stem cells (iPSCs) to be realized, it is necessary to ask if and how efficiently they may be differentiated to functional cells of various lineages. Here, we have directly compared the neural-differentiation capacity of human iPSCs and embryonic stem cells (ESCs). We have shown that human iPSCs use the same transcriptional network to generate neuroepithelia and functionally appropriate neuronal types over the same developmental time course as hESCs in response to the same set of morphogens; however, they do it with significantly reduced efficiency and increased variability. These results were consistent across iPSC lines and independent of the set of reprogramming transgenes used to derive iPSCs as well as the presence or absence of reprogramming transgenes in iPSCs. These findings, which show a need for improving differentiation potency of iPSCs, suggest the possibility of employing human iPSCs in pathological studies, therapeutic screening, and autologous cell transplantation.
Collapse
|
563
|
Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc Natl Acad Sci U S A 2010; 107:4335-40. [PMID: 20160098 DOI: 10.1073/pnas.0910012107] [Citation(s) in RCA: 752] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
For the promise of human induced pluripotent stem cells (iPSCs) to be realized, it is necessary to ask if and how efficiently they may be differentiated to functional cells of various lineages. Here, we have directly compared the neural-differentiation capacity of human iPSCs and embryonic stem cells (ESCs). We have shown that human iPSCs use the same transcriptional network to generate neuroepithelia and functionally appropriate neuronal types over the same developmental time course as hESCs in response to the same set of morphogens; however, they do it with significantly reduced efficiency and increased variability. These results were consistent across iPSC lines and independent of the set of reprogramming transgenes used to derive iPSCs as well as the presence or absence of reprogramming transgenes in iPSCs. These findings, which show a need for improving differentiation potency of iPSCs, suggest the possibility of employing human iPSCs in pathological studies, therapeutic screening, and autologous cell transplantation.
Collapse
|
564
|
Tat PA, Sumer H, Jones KL, Upton K, Verma PJ. The efficient generation of induced pluripotent stem (iPS) cells from adult mouse adipose tissue-derived and neural stem cells. Cell Transplant 2010; 19:525-36. [PMID: 20144262 DOI: 10.3727/096368910x491374] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Ectopic expression of key reprogramming transgenes in somatic cells enables them to adopt the characteristics of pluripotency. Such cells have been termed induced pluripotent stem (iPS) cells and have revolutionized the field of somatic cell reprogramming, as the need for embryonic material is obviated. One of the issues facing both the clinical translation of iPS cell technology and the efficient derivation of iPS cell lines in the research laboratory is choosing the most appropriate somatic cell type for induction. In this study, we demonstrate the direct reprogramming of a defined population of neural stem cells (NSCs) derived from the subventricular zone (SVZ) and adipose tissue-derived cells (ADCs) from adult mice using retroviral transduction of the Yamanaka factors Oct4, Sox2, Klf4, and c-Myc, and compared the results obtained with a mouse embryonic fibroblast (mEF) control. We isolated mEFs, NSCs, and ADCs from transgenic mice, which possess a GFP transgene under control of the Oct4 promoter, and validated GFP expression as an indicator of reprogramming. While transduction efficiencies were not significantly different among the different cell types (mEFs 68.70 +/- 2.62%, ADCs 70.61 +/- 15.4%, NSCs, 68.72 +/- 3%, p = 0.97), the number of GFP-positive colonies and hence the number of reprogramming events was significantly higher for both NSCs (13.50 +/- 4.10 colonies, 0.13 +/- 0.06%) and ADCs (118.20 +/- 38.28 colonies, 1.14 +/- 0.77%) when compared with the mEF control (3.17 +/- 0.29 colonies, 0.03 +/- 0.005%). ADCs were most amenable to reprogramming with an 8- and 38-fold greater reprogramming efficiency than NSCs and mEFs, respectively. Both NSC iPS and ADC iPS cells were demonstrated to express markers of pluripotency and could differentiate to the three germ layers, both in vitro and in vivo, to cells representative of the three germ lineages. Our findings confirm that ADCs are an ideal candidate as a readily accessible somatic cell type for high efficiency establishment of iPS cell lines.
Collapse
Affiliation(s)
- Pollyanna A Tat
- Centre for Reproduction and Development, Monash Institute of Medical Research, Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
565
|
Mason C, Dunnill P. Assessing the value of autologous and allogeneic cells for regenerative medicine. Regen Med 2010; 4:835-53. [PMID: 19903003 DOI: 10.2217/rme.09.64] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The advantages and disadvantages of autologous and allogeneic human cells for regenerative medicine are summarized. The comparison of relative advantages includes: ease and cost of treating large numbers of patients, the speed of availability of therapy and the differing complexity of the development pathways. The comparison of relative disadvantages deals with issues such as variability of source material, the risks of cell abnormality and of viral and prion contamination, and the sensitive issues surrounding use of embryo-derived cells. From the comparisons, several potentially decisive issues are drawn out, such as possible immune response and teratoma formation, the impact of patents and the virtues of hospital versus industry-centered development.
Collapse
Affiliation(s)
- Chris Mason
- Advanced Centre for Biochemical Engineering, University College London, London, UK.
| | | |
Collapse
|
566
|
Lefort N, Perrier AL, Laâbi Y, Varela C, Peschanski M. Human embryonic stem cells and genomic instability. Regen Med 2010; 4:899-909. [PMID: 19903007 DOI: 10.2217/rme.09.63] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Owing to their original properties, pluripotent human embryonic stem cells (hESCs) and their progenies are highly valuable not only for regenerative medicine, but also as tools to study development and pathologies or as cellular substrates to screen and test new drugs. However, ensuring their genomic integrity is one important prerequisite for both research and therapeutic applications. Until recently, several studies about the genomic stability of cultured hESCs had described chromosomal or else large genomic alterations detectable with conventional karyotypic methods. In the past year, several laboratories have reported many small genomic alterations, in the megabase-sized range, using more sensitive karyotyping methods, showing that hESCs are prone to acquire focal genomic abnormalities in culture. As these alterations were found to be nonrandom, these findings strongly advocate for high-resolution monitoring of human pluripotent stem cell lines, especially when intended to be used for clinical applications.
Collapse
Affiliation(s)
- Nathalie Lefort
- Institute for Stem cell Therapy and Exploration of Monogenic diseases, Desbruères, 91030 Evry cedex, France.
| | | | | | | | | |
Collapse
|
567
|
Mayhew CN, Wells JM. Converting human pluripotent stem cells into beta-cells: recent advances and future challenges. Curr Opin Organ Transplant 2010; 15:54-60. [PMID: 19855279 PMCID: PMC2832838 DOI: 10.1097/mot.0b013e3283337e1c] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE OF REVIEW The transplantation of insulin-producing beta-cells derived from human embryonic stem cells and induced pluripotent stem cells (collectively termed pluripotent stem cells or PSCs) holds great promise for therapy of diabetes mellitus. The purpose of this review is to summarize recent advances in this area, emphasizing the importance of studies of endocrine pancreas development in efforts to direct PSC differentiation into endocrine cells, as well as to outline the major challenges remaining before transplantation of PSC-derived beta-cells can become a reality. RECENT FINDINGS Although several protocols to generate glucose-responsive pancreatic beta-cells in vitro have been described, the most successful approaches are those that most closely mimic embryonic development of the endocrine pancreas. Until recently, cells generated by these methods have exhibited immature pancreatic endocrine phenotypes. However, protocols that generate more functional beta-like cells have now been described. In addition, small molecules are being used to improve protocols to direct differentiation of PSCs into endoderm and pancreatic lineages. SUMMARY Advances over the last decade suggest that generating functional beta-cells from human PSCs is achievable. However, there are aspects of beta-cell development that are not well understood and are hampering generation of PSC-derived beta-cells. In particular, the signaling pathways that instruct endocrine progenitor cells to differentiate into mature and functional beta-cells are poorly understood. Other significant obstacles remain, including the need for safe and cost-effective differentiation methods for large-scale generation of transplantation quality beta-cells, methods to prevent immune rejection of grafted tissues, and amelioration of the risks of tumorigenesis.
Collapse
Affiliation(s)
- Christopher N Mayhew
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | |
Collapse
|
568
|
Directed differentiation of hematopoietic precursors and functional osteoclasts from human ES and iPS cells. Blood 2010; 115:2769-76. [PMID: 20065292 DOI: 10.1182/blood-2009-07-234690] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The directed differentiation of human pluripotent stem cells offers the unique opportunity to generate a broad spectrum of human cell types and tissues for transplantation, drug discovery, and studying disease mechanisms. Here, we report the stepwise generation of bone-resorbing osteoclasts from human embryonic and induced pluripotent stem cells. Generation of a primitive streak-like population in embryoid bodies, followed by specification to hematopoiesis and myelopoiesis by vascular endothelial growth factor and hematopoietic cytokines in serum-free media, yielded a precursor population enriched for cells expressing the monocyte-macrophage lineage markers CD14, CD18, CD11b, and CD115. When plated in monolayer culture in the presence of macrophage colony-stimulating factor and receptor activator of nuclear factor-kappaB ligand (RANKL), these precursors formed large, multinucleated osteoclasts that expressed tartrate-resistant acid phosphatase and were capable of resorption. No tartrate-resistant acid phosphatase-positive multinucleated cells or resorption pits were observed in the absence of RANKL. Molecular analyses confirmed the expression of the osteoclast marker genes NFATc1, cathepsin K, and calcitonin receptor in a RANKL-dependent manner, and confocal microscopy demonstrated the coexpression of the alphavbeta3 integrin, cathepsin K and F-actin rings characteristic of active osteoclasts. Generating hematopoietic and osteoclast populations from human embryonic and induced pluripotent stem cells will be invaluable for understanding embryonic bone development and postnatal bone disease.
Collapse
|
569
|
Takahashi K, Narita M, Yokura M, Ichisaka T, Yamanaka S. Human induced pluripotent stem cells on autologous feeders. PLoS One 2009; 4:e8067. [PMID: 19956543 PMCID: PMC2780725 DOI: 10.1371/journal.pone.0008067] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 10/30/2009] [Indexed: 12/31/2022] Open
Abstract
Background For therapeutic usage of induced Pluripotent Stem (iPS) cells, to accomplish xeno-free culture is critical. Previous reports have shown that human embryonic stem (ES) cells can be maintained in feeder-free condition. However, absence of feeder cells can be a hostile environment for pluripotent cells and often results in karyotype abnormalities. Instead of animal feeders, human fibroblasts can be used as feeder cells of human ES cells. However, one still has to be concerned about the existence of unidentified pathogens, such as viruses and prions in these non-autologous feeders. Methodology/Principal Findings This report demonstrates that human induced Pluripotent Stem (iPS) cells can be established and maintained on isogenic parental feeder cells. We tested four independent human skin fibroblasts for the potential to maintain self-renewal of iPS cells. All the fibroblasts tested, as well as their conditioned medium, were capable of maintaining the undifferentiated state and normal karyotypes of iPS cells. Furthermore, human iPS cells can be generated on isogenic parental fibroblasts as feeders. These iPS cells carried on proliferation over 19 passages with undifferentiated morphologies. They expressed undifferentiated pluripotent cell markers, and could differentiate into all three germ layers via embryoid body and teratoma formation. Conclusions/Significance These results suggest that autologous fibroblasts can be not only a source for iPS cells but also be feeder layers. Our results provide a possibility to solve the dilemma by using isogenic fibroblasts as feeder layers of iPS cells. This is an important step toward the establishment of clinical grade iPS cells.
Collapse
Affiliation(s)
- Kazutoshi Takahashi
- Center for iPS cell Research and Application, Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
- * E-mail: (KT); (SY)
| | - Megumi Narita
- Center for iPS cell Research and Application, Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Midori Yokura
- Center for iPS cell Research and Application, Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Tomoko Ichisaka
- Center for iPS cell Research and Application, Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Shinya Yamanaka
- Center for iPS cell Research and Application, Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
- Department of Stem Cell Biology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- Yamanaka iPS Cell Special Project, Japan Science and Technology Agency, Kawaguchi, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, United States of America
- * E-mail: (KT); (SY)
| |
Collapse
|
570
|
Josephson R, Auerbach J. Authentication and banking of human pluripotent stem cells. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2009; Chapter 1:Unit 1C.9. [PMID: 19960451 DOI: 10.1002/9780470151808.sc01c09s11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Pluripotent human stem cell lines from embryos or reprogrammed adult cells are not all alike. Cell lines differ widely in their propensity for differentiation, their chromosomal integrity and epigenetic state, immunological profiles, and their availability for research. It is important that all pluripotent cell lines be protected from loss by being properly banked and authenticated, which will also protect current experimental data by enabling its future reproducibility. This unit considers basic guidelines for banking and authentication of pluripotent stem cells that should be easily implementable within any laboratory. Cell Banking is the disciplined preservation of a cell stock in the originally obtained state, as well as stocks representing the baseline state for experimental efforts. Each of these stocks must be authenticated appropriately. Authentication of pluripotent lines verifies five properties: the unique identity of the line, its sterility or freedom from contaminating microorganisms and pathogens, the integrity and stability of its genome, its expression of typical markers of the stem cell phenotype, and its pluripotency upon differentiation. This unit lists and compares several assays to verify each of these stem cell line properties. Thanks to recent advances in molecular biology and the availability of state-of-the-art assays from service providers, the time and material costs of banking and authentication are not excessive for the typical research laboratory.
Collapse
|
571
|
Lock LT, Tzanakakis ES. Expansion and differentiation of human embryonic stem cells to endoderm progeny in a microcarrier stirred-suspension culture. Tissue Eng Part A 2009; 15:2051-63. [PMID: 19196140 DOI: 10.1089/ten.tea.2008.0455] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Embryonic stem cells (ESCs) with their abilities for extensive proliferation and multi-lineage differentiation can serve as a renewable source of cellular material in regenerative medicine. However, the development of processes for large-scale generation of human ESCs (hESCs) or their progeny will be necessary before hESC-based therapies become a reality. We hypothesized that microcarrier stirred-suspension bioreactors characterized by scalability, straightforward operation, and tight control of the culture environment can be used for hESC culture and directed differentiation. Under appropriate conditions, the concentration of hESCs cultured in a microcarrier bioreactor increased 34- to 45-fold over 8 days. The cells retained the expression of pluripotency markers such as OCT3/4A, NANOG, and SSEA4, as assessed by quantitative PCR, immunocytochemistry, and flow cytometry. We further hypothesized that hESCs on microcarriers can be induced to definitive endoderm (DE) when incubated with physiologically relevant factors. In contrast to embryoid body cultures, all hESCs on microcarriers are exposed to soluble stimuli in the bulk medium facilitating efficient transition to DE. After reaching a peak concentration, hESCs in microcarrier cultures were incubated in medium containing activin A, Wnt3a, and low concentration of serum. More than 80% of differentiated hESCs coexpressed FOXA2 and SOX17 in addition to other DE markers, whereas the expression of non-DE genes was either absent or minimal. We also demonstrate that the hESC-to-DE induction in microcarrier cultures is scalable. Our findings support the use of microcarrier bioreactors for the generation of endoderm progeny from hESCs including pancreatic islets and liver cells in therapeutically useful quantities.
Collapse
Affiliation(s)
- Lye T Lock
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, New York 14260, USA
| | | |
Collapse
|
572
|
Mohr JC, Zhang J, Azarin SM, Soerens AG, de Pablo JJ, Thomson JA, Lyons GE, Palecek SP, Kamp TJ. The microwell control of embryoid body size in order to regulate cardiac differentiation of human embryonic stem cells. Biomaterials 2009; 31:1885-93. [PMID: 19945747 DOI: 10.1016/j.biomaterials.2009.11.033] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2009] [Accepted: 11/13/2009] [Indexed: 12/21/2022]
Abstract
The differentiation of human embryonic stem cells (hESCs) into cardiomyocytes (CMs) using embryoid bodies (EBs) is relatively inefficient and highly variable. Formation of EBs using standard enzymatic disaggregation techniques results in a wide range of sizes and geometries of EBs. Use of a 3-D cuboidal microwell system to culture hESCs in colonies of defined dimensions, 100-500 microm in lateral dimensions and 120 microm in depth, enabled formation of more uniform-sized EBs. The 300 microm microwells produced highest percentage of contracting EBs, but flow cytometry for myosin light chain 2A (MLC2a) expressing cells revealed a similar percentage (approximately 3%) of cardiomyocytes formed in EBs from 100 microm to 300 microm microwells. These data, and immunolabeling with anti-MF20 and MLC2a, suggest that the smaller EBs are less likely to form contracting EBs, but those contracting EBs are relatively enriched in cardiomyocytes compared to larger EB sizes where CMs make up a proportionately smaller fraction of the total cells. We conclude that microwell-engineered EB size regulates cardiogenesis and can be used for more efficient and reproducible formation of hESC-CMs needed for research and therapeutic applications.
Collapse
Affiliation(s)
- Jeffrey C Mohr
- Department of Chemical and Biological Engineering, University of Wisconsin College of Engineering, 1415 Engineering Drive, Madison, WI 53706, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
573
|
Lau F, Ahfeldt T, Osafune K, Akustsu H, Cowan CA. Induced pluripotent stem (iPS) cells: an up-to-the-minute review. F1000 BIOLOGY REPORTS 2009; 1:84. [PMID: 20948605 PMCID: PMC2948253 DOI: 10.3410/b1-84] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Recent advances in nuclear reprogramming technology allow the transformation of terminally differentiated, adult cells into induced pluripotent stem cells whose phenotype is indistinguishable from that of embryonic stem cells. This leap forward enables the creation of patient-specific pluripotent cell lines that carry disease genotypes. These cell lines could be used both as in vitro models for the study of disease and as potential sources of material for cell replacement therapy. Ultimately, a greater understanding of the process by which cellular identity is shaped and altered may allow the generation of particular cell types for the treatment of degenerative disease.
Collapse
Affiliation(s)
- Frank Lau
- Stowers Medical Institute and Harvard Stem Cell Institute, Harvard University, Department of Stem Cell and Regenerative Biology, 185 Cambridge Street CPZN 4234, Boston, MA 02114, USA.
| | | | | | | | | |
Collapse
|
574
|
Ruby KM, Zheng B. Gene targeting in a HUES line of human embryonic stem cells via electroporation. Stem Cells 2009; 27:1496-506. [PMID: 19544466 DOI: 10.1002/stem.73] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Genetic modification is critical for achieving the full potential of human embryonic stem (ES) cells as a tool for therapeutic development and for basic research. Targeted modifications in human ES cells have met with limited success because of the unique culture conditions for many human ES cell lines. The HUES lines of human ES cells were developed for ease of manipulation and are gaining increased utility in stem cell research. We tested conditions for gene targeting via electroporation in the HUES-9 human ES cell line and demonstrate here successful gene targeting at the gene encoding Fezf2 (also known as Fezl), a transcription factor involved in corticospinal neuron development. With a targeting strategy involving positive and negative selection that is applicable to all genes, we observed a gene targeting frequency of approximately 1.5% for Fezf2, a gene not expressed in human ES cells. We found that conditions developed for gene targeting in mouse ES cells can be readily adapted to HUES cells with few key modifications. HUES-9 cells exhibit an intrinsically high efficiency of clonal expansion and sustain electroporation-based gene targeting procedures without any significant loss of pluripotency marker expression or karyotypic stability. Thus, human ES cell lines adapted for enzymatic passage and efficient clonal expansion can be highly amenable to genetic modifications, which will facilitate their application in basic science and clinical development.
Collapse
Affiliation(s)
- Katherine M Ruby
- Biomedical Sciences Graduate Program and Department of Neurosciences, University of California San Diego, School of Medicine, La Jolla, California 92093-0691, USA
| | | |
Collapse
|
575
|
Koch P, Kokaia Z, Lindvall O, Brüstle O. Emerging concepts in neural stem cell research: autologous repair and cell-based disease modelling. Lancet Neurol 2009; 8:819-29. [PMID: 19679274 DOI: 10.1016/s1474-4422(09)70202-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The increasing availability of human pluripotent stem cells provides new prospects for neural-replacement strategies and disease-related basic research. With almost unlimited potential for self-renewal, the use of human embryonic stem cells (ESCs) bypasses the restricted supply and expandability of primary cells that has been a major bottleneck in previous neural transplantation approaches. Translation of developmental patterning and cell-type specification techniques to human ESC cultures enables in vitro generation of various neuronal and glial cell types. The derivation of stably proliferating neural stem cells from human ESCs further facilitates standardisation and circumvents the problem of batch-to-batch variations commonly encountered in "run-through" protocols, which promote terminal differentiation of pluripotent stem cells into somatic cell types without defined intermediate precursor stages. The advent of cell reprogramming offers an opportunity to translate these advances to induced pluripotent stem cells, thereby enabling the generation of neurons and glia from individual patients. Eventually, reprogramming could provide a supply of autologous neural cells for transplantation, and could lead to the establishment of cellular model systems of neurological diseases.
Collapse
Affiliation(s)
- Philipp Koch
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | | | | | | |
Collapse
|
576
|
Freund C, Mummery CL. Prospects for pluripotent stem cell-derived cardiomyocytes in cardiac cell therapy and as disease models. J Cell Biochem 2009; 107:592-9. [PMID: 19449339 DOI: 10.1002/jcb.22164] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The derivation of embryonic stem cells (hESC) from human embryos a decade ago started a new era in perspectives for cell therapy as well as understanding human development and disease. More recently, reprogramming of somatic cells to an embryonic stem cell-like state (induced pluripotent stem cells, iPS) presented a new milestone in this area, making it possible to derive all cells types from any patients bearing specific genetic mutations. With the development of efficient differentiation protocols we are now able to use the derivatives of pluripotent stem cells to study mechanisms of disease and as human models for drug and toxicology testing. In addition derivatives of pluripotent stem cells are now close to be used in clinical practice although for the heart, specific additional challenges have been identified that preclude short-term application in cell therapy. Here we review techniques presently used to induce differentiation of pluripotent stem cells into cardiomyocytes and the potential these cells have as disease models and for therapy.
Collapse
Affiliation(s)
- Christian Freund
- Department of Anatomy & Embryology, Leiden University Medical Center, The Netherlands
| | | |
Collapse
|
577
|
|
578
|
Carpenter MK, Frey-Vasconcells J, Rao MS. Developing safe therapies from human pluripotent stem cells. Nat Biotechnol 2009; 27:606-13. [PMID: 19587662 DOI: 10.1038/nbt0709-606] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
579
|
Borowiak M, Melton DA. How to make beta cells? Curr Opin Cell Biol 2009; 21:727-32. [PMID: 19781928 DOI: 10.1016/j.ceb.2009.08.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 08/25/2009] [Indexed: 02/07/2023]
Abstract
Insulin-producing beta cells are lost or insufficient in diabetic patients, presenting the medical challenge for new beta cells. Currently, there are three strategies that offer promise. One involves the generation of beta cells de novo by directing the differentiation of either embryonic stem cells or induced pluripotent cells to the beta cell lineage. The second is based on the conversion of another terminally differentiated cell to beta cells in a process called reprogramming. The third approach is to promote the replication of existing beta cells either in vivo or in vitro. Significant progress is evident for each strategy, but it remains unclear which approach will ultimately prove successful.
Collapse
Affiliation(s)
- Malgorzata Borowiak
- Harvard Stem Cell Institute, Department of Stem Cell and Regeneration Biology, Harvard University, Cambridge, MA 02138, USA.
| | | |
Collapse
|
580
|
Derivation of insulin-producing cells from human embryonic stem cells. Stem Cell Res 2009; 3:73-87. [DOI: 10.1016/j.scr.2009.08.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 08/01/2009] [Accepted: 08/18/2009] [Indexed: 12/21/2022] Open
|
581
|
|
582
|
Lee H, Park J, Forget BG, Gaines P. Induced pluripotent stem cells in regenerative medicine: an argument for continued research on human embryonic stem cells. Regen Med 2009; 4:759-69. [DOI: 10.2217/rme.09.46] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Human embryonic stem cells (ESCs) can be induced to differentiate into a wide range of tissues that soon could be used for therapeutic applications in regenerative medicine. Despite their developmental potential, sources used to generate human ESC lines raise serious ethical concerns, which recently prompted efforts to reprogram somatic cells back to a pluripotent state. These efforts resulted in the generation of induced pluripotent stem (iPS) cells that are functionally similar to ESCs. However, the genetic manipulations required to generate iPS cells may complicate their growth and developmental characteristics, which poses serious problems in predicting how they will behave when used for tissue-regenerative purposes. In this article we summarize the recently developed methodologies used to generate iPS cells, including those that minimize their genetic manipulation, and discuss several important complicating features of iPS cells that may compromise their future use for therapies in regenerative medicine.
Collapse
Affiliation(s)
- Han Lee
- Yale School of Medicine, Department of Genetics,New Haven, CT 06520, USA
| | - Jung Park
- Kaiser Permanente Los Angeles Medical Center, Department of Internal Medicine, Los Angeles, CA 90027, USA
| | - Bernard G Forget
- Yale School of Medicine, Department of Internal Medicine, New Haven, CT 06520, USA
| | - Peter Gaines
- University of Massachusetts Lowell, Department of Biological Sciences, 515 Olsen Hall, One University Avenue, Lowell, MA 01854, USA
| |
Collapse
|
583
|
Generation of pluripotent stem cells from patients with type 1 diabetes. Proc Natl Acad Sci U S A 2009; 106:15768-73. [PMID: 19720998 DOI: 10.1073/pnas.0906894106] [Citation(s) in RCA: 407] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Type 1 diabetes (T1D) is the result of an autoimmune destruction of pancreatic beta cells. The cellular and molecular defects that cause the disease remain unknown. Pluripotent cells generated from patients with T1D would be useful for disease modeling. We show here that induced pluripotent stem (iPS) cells can be generated from patients with T1D by reprogramming their adult fibroblasts with three transcription factors (OCT4, SOX2, KLF4). T1D-specific iPS cells, termed DiPS cells, have the hallmarks of pluripotency and can be differentiated into insulin-producing cells. These results are a step toward using DiPS cells in T1D disease modeling, as well as for cell replacement therapy.
Collapse
|
584
|
Preynat-Seauve O, Suter DM, Tirefort D, Turchi L, Virolle T, Chneiweiss H, Foti M, Lobrinus JA, Stoppini L, Feki A, Dubois-Dauphin M, Krause KH. Development of human nervous tissue upon differentiation of embryonic stem cells in three-dimensional culture. STEM CELLS (DAYTON, OHIO) 2009; 27:509-20. [PMID: 19074418 DOI: 10.1634/stemcells.2008-0600] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Researches on neural differentiation using embryonic stem cells (ESC) require analysis of neurogenesis in conditions mimicking physiological cellular interactions as closely as possible. In this study, we report an air-liquid interface-based culture of human ESC. This culture system allows three-dimensional cell expansion and neural differentiation in the absence of added growth factors. Over a 3-month period, a macroscopically visible, compact tissue developed. Histological coloration revealed a dense neural-like neural tissue including immature tubular structures. Electron microscopy, immunochemistry, and electrophysiological recordings demonstrated a dense network of neurons, astrocytes, and oligodendrocytes able to propagate signals. Within this tissue, tubular structures were niches of cells resembling germinal layers of human fetal brain. Indeed, the tissue contained abundant proliferating cells expressing markers of neural progenitors. Finally, the capacity to generate neural tissues on air-liquid interface differed for different ESC lines, confirming variations of their neurogenic potential. In conclusion, this study demonstrates in vitro engineering of a human neural-like tissue with an organization that bears resemblance to early developing brain. As opposed to previously described methods, this differentiation (a) allows three-dimensional organization, (b) yields dense interconnected neural tissue with structurally and functionally distinct areas, and (c) is spontaneously guided by endogenous developmental cues.
Collapse
Affiliation(s)
- Olivier Preynat-Seauve
- Department of Pathology and Immunology, University of Geneva, Geneva Hospital, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
585
|
Toward clinical therapies using hematopoietic cells derived from human pluripotent stem cells. Blood 2009; 114:3513-23. [PMID: 19652198 DOI: 10.1182/blood-2009-03-191304] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) provide remarkable cellular platforms to better understand human hematopoiesis and to develop clinically applicable hematopoietic cell-based therapies. Over the past decade, hESCs have been used to characterize molecular and cellular mechanisms underpinning the differentiation of hematopoietic progenitors and mature, functional hematopoietic cells. These advances are now poised to lead to clinical translation of hESC- and iPSC-derived hematopoietic cells for novel therapies in the next few years. On the basis of areas of recent success, initial clinical use of hematopoietic cells derived from human pluripotent stem cells will probably be in the areas of transfusion therapies (erythrocytes and platelets) and immune therapies (natural killer cells). In contrast, efficient development and isolation of hematopoietic stem cells capable of long-term, multilineage engraftment still remains a significant challenge. Technical, safety, and regulatory concerns related to clinical applications of human PSCs must be appropriately addressed. However, proper consideration of these issues should facilitate and not inhibit clinical translation of new therapies. This review outlines the current status of hematopoietic cell development and what obstacles must be surmounted to bring hematopoietic cell therapies from human PSCs from "bench to bedside."
Collapse
|
586
|
Geens M, Mateizel I, Sermon K, De Rycke M, Spits C, Cauffman G, Devroey P, Tournaye H, Liebaers I, Van de Velde H. Human embryonic stem cell lines derived from single blastomeres of two 4-cell stage embryos. Hum Reprod 2009; 24:2709-17. [PMID: 19633307 PMCID: PMC2763129 DOI: 10.1093/humrep/dep262] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Recently, we demonstrated that single blastomeres of a 4-cell stage human embryo are able to develop into blastocysts with inner cell mass and trophectoderm. To further investigate potency at the 4-cell stage, we aimed to derive pluripotent human embryonic stem cells (hESC) from single blastomeres. METHODS Four 4-cell stage embryos were split on Day 2 of preimplantation development and the 16 blastomeres were individually cultured in sequential medium. On Day 3 or 4, the blastomere-derived embryos were plated on inactivated mouse embryonic fibroblasts (MEFs). RESULTS Ten out of sixteen blastomere-derived morulae attached to the MEFs, and two produced an outgrowth. They were mechanically passaged onto fresh MEFs as described for blastocyst ICM-derived hESC, and shown to express the typical stemness markers by immunocytochemistry and/or RT-PCR. In vivo pluripotency was confirmed by the presence of all three germ layers in the teratoma obtained after injection in immunodeficient mice. The first hESC line displays a mosaic normal/abnormal 46, XX, dup(7)(q33qter), del(18)(q23qter) karyotype. The second hESC line displays a normal 46, XY karyotype. CONCLUSION We report the successful derivation and characterization of two hESC lines from single blastomeres of four split 4-cell stage human embryos. These two hESC lines were derived from distinct embryos, proving that at least one of the 4-cell stage blastomeres is pluripotent.
Collapse
Affiliation(s)
- Mieke Geens
- Department of Embryology and Genetics, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
587
|
Chin MH, Mason MJ, Xie W, Volinia S, Singer M, Peterson C, Ambartsumyan G, Aimiuwu O, Richter L, Zhang J, Khvorostov I, Ott V, Grunstein M, Lavon N, Benvenisty N, Croce CM, Clark AT, Baxter T, Pyle AD, Teitell MA, Pelegrini M, Plath K, Lowry WE. Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell Stem Cell 2009; 5:111-23. [PMID: 19570518 PMCID: PMC3448781 DOI: 10.1016/j.stem.2009.06.008] [Citation(s) in RCA: 727] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Revised: 05/30/2009] [Accepted: 06/12/2009] [Indexed: 11/21/2022]
Abstract
Induced pluripotent stem cells (iPSCs) outwardly appear to be indistinguishable from embryonic stem cells (ESCs). A study of gene expression profiles of mouse and human ESCs and iPSCs suggests that, while iPSCs are quite similar to their embryonic counterparts, a recurrent gene expression signature appears in iPSCs regardless of their origin or the method by which they were generated. Upon extended culture, hiPSCs adopt a gene expression profile more similar to hESCs; however, they still retain a gene expression signature unique from hESCs that extends to miRNA expression. Genome-wide data suggested that the iPSC signature gene expression differences are due to differential promoter binding by the reprogramming factors. High-resolution array profiling demonstrated that there is no common specific subkaryotypic alteration that is required for reprogramming and that reprogramming does not lead to genomic instability. Together, these data suggest that iPSCs should be considered a unique subtype of pluripotent cell.
Collapse
Affiliation(s)
- Mark H. Chin
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Mike J. Mason
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Statistics, University of California, Los Angeles, CA 90095, USA
| | - Wei Xie
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Stefano Volinia
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Mike Singer
- Roche NimbleGen, Inc., Madison, WI 53719, USA
| | - Cory Peterson
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Gayane Ambartsumyan
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | - Otaren Aimiuwu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | - Laura Richter
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | - Jin Zhang
- Departments of Pathology and Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Ivan Khvorostov
- Departments of Pathology and Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Vanessa Ott
- Roche NimbleGen, Inc., Madison, WI 53719, USA
| | - Michael Grunstein
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Neta Lavon
- Department of Genetics, Hebrew University, Jerusalem 91904, Israel
| | | | - Carlo M. Croce
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Amander T. Clark
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
- Broad Stem Cell Center, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Tim Baxter
- Roche NimbleGen, Inc., Madison, WI 53719, USA
| | - April D. Pyle
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
- Broad Stem Cell Center, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Mike A. Teitell
- Departments of Pathology and Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Broad Stem Cell Center, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Matteo Pelegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Kathrin Plath
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Broad Stem Cell Center, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - William E. Lowry
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
- Broad Stem Cell Center, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
588
|
Saeki K, Saeki K, Nakahara M, Matsuyama S, Nakamura N, Yogiashi Y, Yoneda A, Koyanagi M, Kondo Y, Yuo A. A feeder-free and efficient production of functional neutrophils from human embryonic stem cells. Stem Cells 2009; 27:59-67. [PMID: 18845766 DOI: 10.1634/stemcells.2007-0980] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A novel, feeder-free hematopoietic differentiation protocol was established for highly efficient production of neutrophils from human embryonic stem cells (hESCs). For the induction of differentiation, spheres were generated in the presence of serum and cytokine cocktail and subjected to attachment culture on gelatin-coated plates. After approximately 2 weeks, a sac-like structure filled with abundant round cells emerged at the center of flattened spheres. After cutting off this sac-like structure, round cells actively proliferated, either floating in the supernatant or associated weakly with the adherent cells. Almost all of these round cells were CD45-positive hematopoietic cells with myeloid phagocytic markers (CD33 and CD11b), and approximately 30%-50% of the round cells were mature neutrophils, as judged from morphology, cytochemical characteristics (myeloperoxidase and neutrophil alkaline phosphatase), and neutrophil-specific cell surface markers (CD66b, CD16b, and GPI-80). In addition, hESC-derived neutrophils had chemotactic capacity in response to the bacterial chemotactic peptide formyl-methionyl-leucyl-phenylalanine and neutrophil-specific chemokine interleukin (IL)-8. Using "semipurified" neutrophils migrated to IL-8, both phagocytic and respiratory burst activities were demonstrated. Finally, it was shown that hESC-derived neutrophils had chemotactic activity in vivo in a murine air-pouch inflammatory model. The present results indicate successful induction of functional mature neutrophils from hESCs via highly efficient feeder-free differentiation culture system of human hematopoietic cells.
Collapse
Affiliation(s)
- Koichi Saeki
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
589
|
Pal R, Totey S, Mamidi MK, Bhat VS, Totey S. Propensity of human embryonic stem cell lines during early stage of lineage specification controls their terminal differentiation into mature cell types. Exp Biol Med (Maywood) 2009; 234:1230-43. [PMID: 19546356 DOI: 10.3181/0901-rm-38] [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: 02/05/2023] Open
Abstract
Human embryonic stem cells (hESCs) are able to stably maintain their characteristics for an unlimited period; nevertheless, substantial differences among cell lines in gene and protein expression not manifested during the undifferentiated state may appear when cells differentiate. It is widely accepted that developing an efficient protocol to control the differentiation of hESCs will enable us to produce adequate numbers of desired cell types with relative ease for diverse applications ranging from basic research to cell therapy and drug screening. Hence of late, there has been considerable interest in understanding whether and how hESC lines are equivalent or different to each other in their in vitro developmental tendencies. In this study, we compared the developmental competences of two hESC lines (HUES-9 and HUES-7) at molecular, cellular and functional levels, upon spontaneous differentiation without any added inducing agents. Both cell lines generated the three embryonic germ layers, extra-embryonic tissues and primordial germ cells during embryoid body (EB) formation. However HUES-9 showed a stronger propensity towards formation of neuroectodermal lineages, whereas HUES-7 differentiated preferentially into mesoderm and endoderm. Upon further differentiation, HUES-9 generated largely neural cells (neurons, oligodendrocytes, astrocytes and gangliosides) whereas HUES-7 formed mesendodermal derivatives, including cardiomyocytes, skeletal myocytes, endothelial cells, hepatocytes and pancreatic cells. Overall, our findings endorse the hypothesis that independently-derived hESCs biologically differ among themselves, thereby displaying varying differentiation propensity. These subtle differences not only highlight the importance of screening and deriving lines for lineage-specific differentiation but also indicate that individual lines may possess a repertoire of capabilities that is unique.
Collapse
Affiliation(s)
- Rajarshi Pal
- Manipal Institute of Regenerative Medicine, Manipal University Branch Campus, Bangalore 560071, India
| | | | | | | | | |
Collapse
|
590
|
Abstract
We review progress towards the goal of utilizing stem cells as a source of engineered pancreatic beta-cells for therapy of diabetes. Protocols for the in vitro differentiation of embryonic stem (ES) cells based on normal developmental cues have generated beta-like cells that produce high levels of insulin, albeit at low efficiency and without full responsiveness to extracellular levels of glucose. Induced pluripotent stem (iPS) cells also can yield insulin-producing cells following similar approaches. An important recent report shows that when transplanted into mice, human ES-derived cells with a phenotype corresponding to pancreatic endoderm matured to yield cells capable of maintaining near-normal regulation of blood sugar [Kroon et al., 2008]. Major hurdles that must be overcome to enable the broad clinical translation of these advances include teratoma formation by ES and iPS cells, and the need for immunosuppressive drugs. Classes of stem cells that can be expanded extensively in culture but do not form teratomas, such as amniotic fluid-derived stem cells and hepatic stem cells, offer possible alternatives for the production of beta-like cells, but further evidence is required to document this potential. Generation of autologous iPS cells should prevent transplant rejection, but may prove prohibitively expensive. Banking strategies to identify small numbers of stem cell lines homozygous for major histocompatibility loci have been proposed to enable beneficial genetic matching that would decrease the need for immunosuppression.
Collapse
Affiliation(s)
- Mark E Furth
- Department of Urology and Wake Forest, Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | |
Collapse
|
591
|
Kumar M, Kaushalya SK, Gressens P, Maiti S, Mani S. Optimized Derivation and Functional Characterization of 5-HT Neurons from Human Embryonic Stem Cells. Stem Cells Dev 2009; 18:615-27. [DOI: 10.1089/scd.2008.0181] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Manoj Kumar
- Department of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| | | | - Pierre Gressens
- Inserm, U676, Paris, France
- Université Paris 7, Faculté de Médecine Denis Diderot, IFR02 and IFR25, Paris, France
| | - Sudipta Maiti
- Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Shyamala Mani
- Department of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| |
Collapse
|
592
|
Abstract
Telomeric regions are known to be transcribed in several organisms. Although originally reported to be transcribed from all chromosomes with enrichment near the inactive X of female cells, we show that telomeric RNAs in fact are enriched on both sex chromosomes of the mouse in a developmentally specific manner. In female stem cells, both active Xs are marked by the RNAs. In male stem cells, both the X and the Y accumulate telomeric RNA. Distribution of telomeric RNAs changes during cell differentiation, after which they associate only with the heterochromatic sex chromosomes of each sex. FISH mapping suggests that accumulated telomeric RNAs localize at the distal telomeric end. Interestingly, telomeric expression changes in cancer and during cellular stress. Furthermore, RNA accumulation increases in Dicer-deficient stem cells, suggesting direct or indirect links to RNAi. We propose that telomeric RNAs are tied to cell differentiation and may be used to mark pluripotency and disease.
Collapse
|
593
|
Borowiak M, Maehr R, Chen S, Chen AE, Tang W, Fox JO, Schreiber SL, Melton DA. Small molecules efficiently direct endodermal differentiation of mouse and human embryonic stem cells. Cell Stem Cell 2009; 4:348-58. [PMID: 19341624 PMCID: PMC4564293 DOI: 10.1016/j.stem.2009.01.014] [Citation(s) in RCA: 326] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 12/02/2008] [Accepted: 01/16/2009] [Indexed: 12/14/2022]
Abstract
An essential step for therapeutic and research applications of stem cells is the ability to differentiate them into specific cell types. Endodermal cell derivatives, including lung, liver, and pancreas, are of interest for regenerative medicine, but efforts to produce these cells have been met with only modest success. In a screen of 4000 compounds, two cell-permeable small molecules were indentified that direct differentiation of ESCs into the endodermal lineage. These compounds induce nearly 80% of ESCs to form definitive endoderm, a higher efficiency than that achieved by Activin A or Nodal, commonly used protein inducers of endoderm. The chemically induced endoderm expresses multiple endodermal markers, can participate in normal development when injected into developing embryos, and can form pancreatic progenitors. The application of small molecules to differentiate mouse and human ESCs into endoderm represents a step toward achieving a reproducible and efficient production of desired ESC derivatives.
Collapse
Affiliation(s)
- Malgorzata Borowiak
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - René Maehr
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Shuibing Chen
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Alice E. Chen
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Weiping Tang
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142, USA
| | - Julia O. Fox
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Stuart L. Schreiber
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Douglas A. Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| |
Collapse
|
594
|
Karumbayaram S, Novitch BG, Patterson M, Umbach JA, Richter L, Lindgren A, Conway A, Clark A, Goldman SA, Plath K, Wiedau-Pazos M, Kornblum HI, Lowry WE. Directed differentiation of human-induced pluripotent stem cells generates active motor neurons. Stem Cells 2009; 27:806-11. [PMID: 19350680 PMCID: PMC2895909 DOI: 10.1002/stem.31] [Citation(s) in RCA: 280] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The potential for directed differentiation of human-induced pluripotent stem (iPS) cells to functional postmitotic neuronal phenotypes is unknown. Following methods shown to be effective at generating motor neurons from human embryonic stem cells (hESCs), we found that once specified to a neural lineage, human iPS cells could be differentiated to form motor neurons with a similar efficiency as hESCs. Human iPS-derived cells appeared to follow a normal developmental progression associated with motor neuron formation and possessed prototypical electrophysiological properties. This is the first demonstration that human iPS-derived cells are able to generate electrically active motor neurons. These findings demonstrate the feasibility of using iPS-derived motor neuron progenitors and motor neurons in regenerative medicine applications and in vitro modeling of motor neuron diseases.
Collapse
Affiliation(s)
- S Karumbayaram
- Department of Neurology, David Geffen School of Medicine at UCLA
| | - BG Novitch
- Department of Neurobiology, David Geffen School of Medicine at UCLA
- Broad Stem Cell Center, UCLA
| | - M Patterson
- Department of Molecular, Cell and Developmental Biology, UCLA
| | - JA Umbach
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA
| | - L Richter
- Department of Molecular, Cell and Developmental Biology, UCLA
| | - A Lindgren
- Department of Molecular, Cell and Developmental Biology, UCLA
| | - A Conway
- Department of Molecular, Cell and Developmental Biology, UCLA
| | - A Clark
- Broad Stem Cell Center, UCLA
- Department of Molecular, Cell and Developmental Biology, UCLA
| | - SA Goldman
- Center for Translational Neuromedicine, Dept. of Neurology, University of Rochester, Rochester, NY
| | - K Plath
- Broad Stem Cell Center, UCLA
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
| | - M Wiedau-Pazos
- Department of Neurology, David Geffen School of Medicine at UCLA
| | - HI Kornblum
- Broad Stem Cell Center, UCLA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA
- The Semel Institute and the Mental Retardation Research Center at UCLA
| | - WE Lowry
- Broad Stem Cell Center, UCLA
- Department of Molecular, Cell and Developmental Biology, UCLA
| |
Collapse
|
595
|
Kumar N, Hinduja I, Nagvenkar P, Pillai L, Zaveri K, Mukadam L, Telang J, Desai S, Mangoli V, Mangoli R, Padgaonkar S, Kaur G, Puri C, Bhartiya D. Derivation and Characterization of Two Genetically Unique Human Embryonic Stem Cell Lines on In-House–Derived Human Feeders. Stem Cells Dev 2009; 18:435-45. [DOI: 10.1089/scd.2008.0234] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Neeraj Kumar
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | | | - Punam Nagvenkar
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | - Lakshmi Pillai
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | | | | | - Jyoti Telang
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | | | | | | | | | - Gurvinder Kaur
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | - Chander Puri
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | - Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
596
|
A small molecule that directs differentiation of human ESCs into the pancreatic lineage. Nat Chem Biol 2009; 5:258-65. [PMID: 19287398 DOI: 10.1038/nchembio.154] [Citation(s) in RCA: 360] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 02/10/2009] [Indexed: 02/06/2023]
Abstract
Stepwise differentiation from embryonic stem cells (ESCs) to functional insulin-secreting beta cells will identify key steps in beta-cell development and may yet prove useful for transplantation therapy for diabetics. An essential step in this schema is the generation of pancreatic progenitors--cells that express Pdx1 and produce all the cell types of the pancreas. High-content chemical screening identified a small molecule, (-)-indolactam V, that induces differentiation of a substantial number of Pdx1-expressing cells from human ESCs. The Pdx1-expressing cells express other pancreatic markers and contribute to endocrine, exocrine and duct cells, in vitro and in vivo. Further analyses showed that (-)-indolactam V works specifically at one stage of pancreatic development, inducing pancreatic progenitors from definitive endoderm. This study describes a chemical screening platform to investigate human ESC differentiation and demonstrates the generation of a cell population that is a key milepost on the path to making beta cells.
Collapse
|
597
|
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: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 02/19/2009] [Indexed: 02/07/2023]
|
598
|
Nakahara M, Saeki K, Nakamura N, Matsuyama S, Yogiashi Y, Yasuda K, Kondo Y, Yuo A. Human Embryonic Stem Cells with Maintenance under a Feeder-Free and Recombinant Cytokine-Free Condition. CLONING AND STEM CELLS 2009; 11:5-18. [DOI: 10.1089/clo.2008.0043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Masako Nakahara
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - Kumiko Saeki
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - Naoko Nakamura
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - Satoko Matsuyama
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - Yoshiko Yogiashi
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - Kazuki Yasuda
- Department of Metabolic Disorder, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | - Yasushi Kondo
- Advanced Medical Research Laboratories, Mitsubishi Tanabe Pharma Corporation, Osaka, Japan
| | - Akira Yuo
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| |
Collapse
|
599
|
Damoiseaux R, Sherman SP, Alva JA, Peterson C, Pyle AD. Integrated chemical genomics reveals modifiers of survival in human embryonic stem cells. Stem Cells 2009; 27:533-542. [PMID: 19074420 PMCID: PMC3962308 DOI: 10.1634/stemcells.2008-0596] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Understanding how survival is regulated in human embryonic stem cells (hESCs) could improve expansion of stem cells for production of cells for regenerative therapy. There is great variability in comparing the differentiation potential of multiple hESC lines. One reason for this is poor survival upon dissociation, which limits selection of homogeneous populations of cells. Understanding the complexity of survival signals has been hindered by the lack of a reproducible system to identify modulators of survival in pluripotent cells. We therefore developed a high-content screening approach with small molecules to examine hESC survival. We have identified novel small molecules that improve survival by inhibiting either Rho-kinase or protein kinase C. Importantly, small molecule targets were verified using short hairpin RNA. Rescreening with stable hESCs that were genetically altered to have increased survival enabled us to identify groups of pathway targets that are important for modifying survival. Understanding how survival is regulated in hESCs could overcome severe technical difficulties in the field, namely expansion of stem cells to improve production of cells and tissues for regenerative therapy.
Collapse
Affiliation(s)
- Robert Damoiseaux
- Molecular Screening Shared Resource, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - Sean P. Sherman
- Molecular Biology Institute, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - Jackelyn A. Alva
- Department of Microbiology, Immunology, and Molecular Genetics, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - Cory Peterson
- Department of Microbiology, Immunology, and Molecular Genetics, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - April D. Pyle
- Molecular Biology Institute, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
- Department of Microbiology, Immunology, and Molecular Genetics, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
600
|
Preynat-Seauve O, Suter DM, Tirefort D, Turchi L, Virolle T, Chneiweiss H, Foti M, Lobrinus JA, Stoppini L, Feki A, Dubois-Dauphin M, Krause KH. Development of human nervous tissue upon differentiation of embryonic stem cells in three-dimensional culture. Stem Cells 2009. [DOI: 10.1002/stem.20080600] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|