451
|
Genbacev O, Krtolica A, Zdravkovic T, Brunette E, Powell S, Nath A, Caceres E, McMaster M, McDonagh S, Li Y, Mandalam R, Lebkowski J, Fisher SJ. Serum-free derivation of human embryonic stem cell lines on human placental fibroblast feeders. Fertil Steril 2005; 83:1517-29. [PMID: 15866593 DOI: 10.1016/j.fertnstert.2005.01.086] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2004] [Revised: 01/24/2005] [Accepted: 01/24/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To derive new human embryonic stem cell (hESC) lines on pathogen-free human placental fibroblast feeders under serum-free conditions. Because the embryo develops in close contact with extraembryonic membranes, we hypothesized that placental mesenchyme might replicate the stem cell niche in situ. DESIGN We isolated and characterized human placental fibroblast lines from individual donors and tested their ability to support growth of federally registered hESC lines. Moreover, we performed extensive pathogen testing to ensure their suitability as feeders for the derivation of therapy-grade hESCs. RESULT(S) Human placental fibroblasts were comparable or superior to mouse embryo fibroblasts as hESC feeders. We used these qualified placental fibroblasts to derive two new hESC lines in knockout Dulbecco's modified Eagle's medium with serum-free 20% knockout serum replacement. The cells, which had a normal karyotype, were grown for more than 25 passages, expressed markers of stemness including Oct-3/4, Tra 1-60, Tra 1-80, and SSEA-4, exhibited high telomerase activity, and differentiated in vitro and in vivo into cells derived from all three germ layers, confirming their pluripotency. Additionally, newly derived hESCs were adapted to growth on a human placental laminin substrate in a defined medium. CONCLUSION(S) To our knowledge, this is the first report of hESC derivation in the absence of serum on qualified pathogen-free human feeders.
Collapse
Affiliation(s)
- Olga Genbacev
- Department of Cell and Tissue Biology, Program in Human Stem Cell Biology, and Developmental and Stem Cell Biology Program, University of California San Francisco, San Francisco, California 94143-0512, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
452
|
Abstract
The application of stem cells in regenerative and reparative therapies is emerging in surgery. Published information can lead to an over simplified view of stem cells with respect to their definitions, tissues of origin, abilities to differentiate into tissue lineages, and their capacity for functional tissue regeneration. The goals of this review article are to define embryonic and adult stem cells, compare differences between them, and summarize their potential clinical applications.
Collapse
Affiliation(s)
- Lisa A Fortier
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
453
|
Bryja V, Bonilla S, Cajánek L, Parish CL, Schwartz CM, Luo Y, Rao MS, Arenas E. An efficient method for the derivation of mouse embryonic stem cells. Stem Cells 2005; 24:844-9. [PMID: 16339993 DOI: 10.1634/stemcells.2005-0444] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mouse embryonic stem cells (mESCs) represent a unique tool for many researchers; however, the process of ESC derivation is often very inefficient and requires high specialization, training, and expertise. To circumvent these limitations, we aimed to develop a simple and efficient protocol based on the use of commercially available products. Here, we present an optimized protocol that we successfully applied to derive ESCs from several knockout mouse strains (Wnt-1, Wnt-5a, Lrp6, and parkin) with 50%-75% efficiency. The methodology is based on the use of mouse embryonic fibroblast feeders, knockout serum replacement (SR), and minimal handling of the blastocyst. In this protocol, all centrifugation steps (as well as the use of trypsin inhibitor) were avoided and replaced by an ESC medium containing fetal calf serum (FCS) after the trypsinizations. We define the potential advantages and disadvantages of using SR and FCS in individual steps of the protocol. We also characterize the ESCs for the expression of ESC markers by immunohistochemistry, Western blot, and a stem cell focused microarray. In summary, we provide a simplified and improved protocol to derive mESCs that can be useful for laboratories aiming to isolate transgenic mESCs for the first time.
Collapse
Affiliation(s)
- Vítezlav Bryja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet S-171 77, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
454
|
Cai J, Chen J, Liu Y, Miura T, Luo Y, Loring JF, Freed WJ, Rao MS, Zeng X. Assessing self-renewal and differentiation in human embryonic stem cell lines. Stem Cells 2005; 24:516-30. [PMID: 16293578 PMCID: PMC1855239 DOI: 10.1634/stemcells.2005-0143] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Like other cell populations, undifferentiated human embryonic stem cells (hESCs) express a characteristic set of proteins and mRNA that is unique to the cells regardless of culture conditions, number of passages, and methods of propagation. We sought to identify a small set of markers that would serve as a reliable indicator of the balance of undifferentiated and differentiated cells in hESC populations. Markers of undifferentiated cells should be rapidly downregulated as the cells differentiate to form embryoid bodies (EBs), whereas markers that are absent or low during the undifferentiated state but that are induced as hESCs differentiate could be used to assess the presence of differentiated cells in the cultures. In this paper, we describe a list of markers that reliably distinguish undifferentiated and differentiated cells. An initial list of approximately 150 genes was generated by scanning published massively parallel signature sequencing, expressed sequence tag scan, and microarray datasets. From this list, a subset of 109 genes was selected that included 55 candidate markers of undifferentiated cells, 46 markers of hESC derivatives, four germ cell markers, and four trophoblast markers. Expression of these candidate marker genes was analyzed in undifferentiated hESCs and differentiating EB populations in four different lines by immunocytochemistry, reverse transcription-polymer-ase chain reaction (RT-PCR), microarray analysis, and quantitative RT-PCR (qPCR). We show that qPCR, with as few as 12 selected genes, can reliably distinguish differentiated cells from undifferentiated hESC populations.
Collapse
Affiliation(s)
- Jingli Cai
- Laboratory of Neurosciences, National Institute on Aging, Department of Health and Human Services (DHHS), Baltimore, MD
| | - Jia Chen
- Cellular Neurobiology Branch, National Institute on Drug Abuse, DHHS, Baltimore, MD
| | - Ying Liu
- Laboratory of Neurosciences, National Institute on Aging, Department of Health and Human Services (DHHS), Baltimore, MD
| | - Takumi Miura
- Laboratory of Neurosciences, National Institute on Aging, Department of Health and Human Services (DHHS), Baltimore, MD
| | - Yongquan Luo
- Laboratory of Neurosciences, National Institute on Aging, Department of Health and Human Services (DHHS), Baltimore, MD
| | - Jeanne F. Loring
- Program in Stem Cells and Regeneration, The Burnham Institute, La Jolla, CA
| | - William J Freed
- Cellular Neurobiology Branch, National Institute on Drug Abuse, DHHS, Baltimore, MD
| | - Mahendra S Rao
- Laboratory of Neurosciences, National Institute on Aging, Department of Health and Human Services (DHHS), Baltimore, MD
| | - Xianmin Zeng
- Cellular Neurobiology Branch, National Institute on Drug Abuse, DHHS, Baltimore, MD
- Buck Institute for Age Research, 8001 Redwood Blvd, Novato, CA
| |
Collapse
|
455
|
Wang L, Menendez P, Cerdan C, Bhatia M. Hematopoietic development from human embryonic stem cell lines. Exp Hematol 2005; 33:987-96. [PMID: 16140146 DOI: 10.1016/j.exphem.2005.06.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The most common human cell-based therapy applied today is hematopoietic stem cell (HSC) transplantation. Currently, human bone marrow, mobilized peripheral blood, and umbilical cord blood represent the major sources of transplantable HSCs, but their availability for use is limited by both compatibility between donor and recipient and required quantity. Although increasing evidence suggests that somatic HSCs can be expanded to meet current needs, their in vivo potential is concomitantly compromised after ex vivo culture. In contrast, human embryonic stem cells (hESC) possess indefinite proliferative capacity in vitro and have been shown to differentiate into the hematopoietic cell fate, giving rise to erythroid, myeloid, and lymphoid lineages using a variety of differentiation procedures. Human ESC-derived hematopoietic cells emerge from a subset of embryonic endothelium expressing PECAM-1, Flk-1, and VE-Cadherin, but lacking CD45 (CD45negPFV). These CD45negPFV precursors are exclusively responsible for hematopoietic potential of differentiated hESCs. hESC-derived hematopoietic cells show similar clonogenic capacity and primitive phenotype to somatic sources of hematopoietic progenitors and possess limited in vivo repopulating capacity in immunodeficient mice, suggestive of HSC function. Here, we will review current progress in studies of hESC-derived hematopoietic cells and discuss the potential precincts and applications.
Collapse
Affiliation(s)
- Lisheng Wang
- Krembil Center for Stem Cell Biology and Regenerative Medicine, Robarts Research Institute, London, Ontario, Canada
| | | | | | | |
Collapse
|
456
|
Noaksson K, Zoric N, Zeng X, Rao MS, Hyllner J, Semb H, Kubista M, Sartipy P. Monitoring Differentiation of Human Embryonic Stem Cells Using Real-Time PCR. Stem Cells 2005; 23:1460-7. [PMID: 16081663 DOI: 10.1634/stemcells.2005-0093] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is a general lack of rapid, sensitive, and quantitative methods for the detection of differentiating human embryonic stem cells (hESCs). Using light microscopy and immunohistochemistry, we observed that morphological changes of differentiating hESCs precede any major alterations in the expression of several commonly used hESC markers (SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, Oct-4, and Nanog). In an attempt to quantify the changes during stochastic differentiation of hESCs, we developed a robust and sensitive multi-marker quantitative real-time polymerase chain reaction (QPCR) method. To maximize the sensitivity of the method, we measured the expression of up- and downregulated genes before and after differentiation of the hESCs. Out of the 12 genes assayed, we found it clearly sufficient to determine the relative differentiation state of the cells by calculating a collective expression index based on the mRNA levels of Oct-4, Nanog, Cripto, and alpha-fetoprotein. We evaluated the method using different hESC lines maintained in either feeder-dependent or feeder-free culture conditions. The QPCR method is very flexible, and by appropriately selecting reporter genes, the method can be designed for various applications. The combination of QPCR with hESC-based technologies opens novel avenues for high-throughput analysis of hESCs in, for example, pharmacological and cytotoxicity screening.
Collapse
Affiliation(s)
- Karin Noaksson
- Cellartis AB, Arvid Wallgrens Backe 20, 413 46 Göteborg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
457
|
Bhattacharya B, Cai J, Luo Y, Miura T, Mejido J, Brimble SN, Zeng X, Schulz TC, Rao MS, Puri RK. Comparison of the gene expression profile of undifferentiated human embryonic stem cell lines and differentiating embryoid bodies. BMC DEVELOPMENTAL BIOLOGY 2005; 5:22. [PMID: 16207381 PMCID: PMC1260016 DOI: 10.1186/1471-213x-5-22] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Accepted: 10/05/2005] [Indexed: 12/26/2022]
Abstract
Background The identification of molecular pathways of differentiation of embryonic stem cells (hESC) is critical for the development of stem cell based medical therapies. In order to identify biomarkers and potential regulators of the process of differentiation, a high quality microarray containing 16,659 seventy base pair oligonucleotides was used to compare gene expression profiles of undifferentiated hESC lines and differentiating embryoid bodies. Results Previously identified "stemness" genes in undifferentiated hESC lines showed down modulation in differentiated cells while expression of several genes was induced as cells differentiated. In addition, a subset of 194 genes showed overexpression of greater than ≥ 3 folds in human embryoid bodies (hEB). These included 37 novel and 157 known genes. Gene expression was validated by a variety of techniques including another large scale array, reverse transcription polymerase chain reaction, focused cDNA microarrays, massively parallel signature sequencing (MPSS) analysis and immunocytochemisty. Several novel hEB specific expressed sequence tags (ESTs) were mapped to the human genome database and their expression profile characterized. A hierarchical clustering analysis clearly depicted a distinct difference in gene expression profile among undifferentiated and differentiated hESC and confirmed that microarray analysis could readily distinguish them. Conclusion These results present a detailed characterization of a unique set of genes, which can be used to assess the hESC differentiation.
Collapse
Affiliation(s)
- Bhaskar Bhattacharya
- Laboratory of Molecular Tumor Biology, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | - Jingli Cai
- Laboratory of Neuroscience, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Youngquan Luo
- Laboratory of Neuroscience, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Takumi Miura
- Laboratory of Neuroscience, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Josef Mejido
- Laboratory of Molecular Tumor Biology, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | - Xianmin Zeng
- National Institute of Drug Abuse, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Mahendra S Rao
- Laboratory of Molecular Tumor Biology, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
- Laboratory of Neuroscience, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Raj K Puri
- Laboratory of Molecular Tumor Biology, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| |
Collapse
|
458
|
Sasaki E, Hanazawa K, Kurita R, Akatsuka A, Yoshizaki T, Ishii H, Tanioka Y, Ohnishi Y, Suemizu H, Sugawara A, Tamaoki N, Izawa K, Nakazaki Y, Hamada H, Suemori H, Asano S, Nakatsuji N, Okano H, Tani K. Establishment of Novel Embryonic Stem Cell Lines Derived from the Common Marmoset (Callithrix jacchus). Stem Cells 2005; 23:1304-13. [PMID: 16109758 DOI: 10.1634/stemcells.2004-0366] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The successful establishment of human embryonic stem cell (hESC) lines has inaugurated a new era in regenerative medicine by facilitating the transplantation of differentiated ESCs to specific organs. However, problems with the safety and efficacy of hESC therapy in vivo remain to be resolved. Preclinical studies using animal model systems, including nonhuman primates, are essential to evaluate the safety and efficacy of hESC therapies. Previously, we demonstrated that common marmosets are suitable laboratory animal models for preclinical studies of hematopoietic stem cell therapies. As this animal model is also applicable to preclinical trials of ESC therapies, we have established novel common marmoset ESC (CMESC) lines. To obtain marmoset embryos, we developed a new embryo collection system, in which blastocysts can be obtained every 3 weeks from each marmoset pair. The inner cell mass was isolated by immunosurgery and plated on a mouse embryonic feeder layer. Some of the CMESC lines were cultured continuously for more than 1 year. These CMESC lines showed alkaline phosphatase activity and expressed stage-specific embryonic antigen (SSEA)-3, SSEA-4, TRA-1-60, and TRA-1-81. On the other hand, SSEA-1 was not detected. Furthermore, our novel CMESCs are pluripotent, as evidenced by in vivo teratoma formation in immunodeficient mice and in vitro differentiation experiments. Our established CMESC lines and the common marmoset provide an excellent experimental model system for understanding differentiation mechanisms, as well as the development of regenerative therapies using hESCs.
Collapse
Affiliation(s)
- Erika Sasaki
- Division of Laboratory Animal Science, Central Institute for Experimental Animals, Kanagawa, and Department of Urology, Urayasu Hospital, Juntendo University, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
459
|
Okamura K, Asahina K, Fujimori H, Ozeki R, Shimizu-Saito K, Tanaka Y, Teramoto K, Arii S, Takase K, Kataoka M, Soeno Y, Tateno C, Yoshizato K, Teraoka H. Generation of hybrid hepatocytes by cell fusion from monkey embryoid body cells in the injured mouse liver. Histochem Cell Biol 2005; 125:247-57. [PMID: 16195892 DOI: 10.1007/s00418-005-0065-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2005] [Indexed: 01/09/2023]
Abstract
Monkey embryonic stem (ES) cells have characteristics that are similar to human ES cells, and might be useful as a substitute model for preclinical research. When embryoid bodies (EBs) formed from monkey ES cells were cultured, expression of many hepatocyte-related genes including cytochrome P450 (Cyp) 3a and Cyp7a1 was observed. Hepatocytes were immunocytochemically observed using antibodies against albumin (ALB), cytokeratin-8/18, and alpha1-antitrypsin in the developing EBs. The in vitro differentiation potential of monkey ES cells into the hepatic lineage prompted us to examine the transplantability of monkey EB cells. As an initial approach to assess the repopulation potential, we transplanted EB cells into immunodeficient urokinase-type plasminogen activator transgenic mice that undergo liver failure. After transplantation, the hepatocyte colonies expressing monkey ALB were observed in the mouse liver. Fluorescence in-situ hybridization revealed that the repopulating hepatocytes arise from cell fusion between transplanted monkey EB cells and recipient mouse hepatocytes. In contrast, neither cell fusion nor repopulation of hepatocytes was observed in the recipient liver after undifferentiated ES cell transplantation. These results indicate that the differentiated cells in developing monkey EBs, but not contaminating ES cells, generate functional hepatocytes by cell fusion with recipient mouse hepatocytes, and repopulate injured mouse liver.
Collapse
Affiliation(s)
- Kentaro Okamura
- Department of Pathological Biochemistry, Medical Research Institute, Tokyo Medical and Dental University, 2-3-10 Kandasurugadai, Chiyoda-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
460
|
Wei CL, Miura T, Robson P, Lim SK, Xu XQ, Lee MYC, Gupta S, Stanton L, Luo Y, Schmitt J, Thies S, Wang W, Khrebtukova I, Zhou D, Liu ET, Ruan YJ, Rao M, Lim B. Transcriptome profiling of human and murine ESCs identifies divergent paths required to maintain the stem cell state. Stem Cells 2005; 23:166-85. [PMID: 15671141 DOI: 10.1634/stemcells.2004-0162] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human embryonic stem cells (hESCs) are an important source of stem cells in regenerative medicine, and much remains unknown about their molecular characteristics. To develop a detailed genomic profile of ESC lines in two different species, we compared transcriptomes of one murine and two different hESC lines by massively parallel signature sequencing (MPSS). Over 2 million signature tags from each line and their differentiating embryoid bodies were sequenced. Major differences and conserved similarities between species identified by MPSS were validated by reverse transcription polymerase chain reaction (RT-PCR) and microarray. The two hESC lines were similar overall, with differences that are attributable to alleles and propagation. Human-mouse comparisons, however, identified only a small (core) set of conserved genes that included genes known to be important in ESC biology, as well as additional novel genes. Identified were major differences in leukemia inhibitory factor, transforming growth factor-beta, and Wnt and fibroblast growth factor signaling pathways, as well as the expression of genes encoding metabolic, cytoskeletal, and matrix proteins, many of which were verified by RT-PCR or by comparing them with published databases. The study reported here underscores the importance of cross-species comparisons and the versatility and sensitivity of MPSS as a powerful complement to current array technology.
Collapse
Affiliation(s)
- Chia Lin Wei
- Genome Institute of Singapore, 60 Biopolis Street, Genome#02-01, Singapore 138672
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
461
|
Western P, Maldonado-Saldivia J, van den Bergen J, Hajkova P, Saitou M, Barton S, Surani MA. Analysis of Esg1 expression in pluripotent cells and the germline reveals similarities with Oct4 and Sox2 and differences between human pluripotent cell lines. Stem Cells 2005; 23:1436-42. [PMID: 16166252 DOI: 10.1634/stemcells.2005-0146] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Establishment of pluripotent epiblast cells is a critical event during early mammalian development because all somatic lineages and the primordial germ cells (PGCs) are derived from them. The epiblast and PGCs are in turn the precursors of pluripotent embryonic stem cells and embryonic germ cells, respectively. Although PGCs are specialized cells, they express several key pluripotency-related genes, such as Oct4 and Sox2. We have analyzed Esg1 expression in mouse and human cells and shown that in the mouse the gene is specifically expressed in preimplantation embryos, stem cells, and the germline. Moreover, Esg1 coexpresses with Oct4 and Sox2, confirming its identity as a marker of the pluripotent cycle. Esg1 is also expressed with Oct4 and Sox2 by human embryonic stem cells and in germ cell carcinoma tissue but not by all human embryonal carcinoma cell lines. These data suggest that together with Oct4 and Sox2, Esg1 plays a conserved role in the pluripotent pathway of mouse and human stem and germ cells.
Collapse
Affiliation(s)
- Patrick Western
- ARC Centre of Excellence in Biotechnology and Development and Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
462
|
Hatzfeld A, Peiffer I, Hatzfeld J. [Human embryonic stem cells: practical problems and scientific potentialities]. ACTA ACUST UNITED AC 2005; 54:94-9. [PMID: 16140469 DOI: 10.1016/j.patbio.2005.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Accepted: 06/20/2005] [Indexed: 11/19/2022]
Abstract
Human embryonic stem cells (hESCs) are derived from pre-implantation embryos given to research with the informed consent of the parents. These cells cannot give rise to a human being: they are not totipotent. They have an unlimited self-renewal capacity and they can generate the three embryonic germ layers, their respective derivatives and the extra-embryonic tissues: they are pluripotent. These cells represent an outstanding material for understanding functional genomics of not yet characterized human genes. They will be an important tool for pharmaceutical and clinical research.
Collapse
Affiliation(s)
- A Hatzfeld
- Centre national de la recherche scientifique, UPR 9045 oncologie virale, institut André-Lwoff, 7, rue Guy-Moquet, 94800 Villejuif, France.
| | | | | |
Collapse
|
463
|
Forrai A, Boyle K, Hart AH, Hartley L, Rakar S, Willson TA, Simpson KM, Roberts AW, Alexander WS, Voss AK, Robb L. Absence of suppressor of cytokine signalling 3 reduces self-renewal and promotes differentiation in murine embryonic stem cells. Stem Cells 2005; 24:604-14. [PMID: 16123385 DOI: 10.1634/stemcells.2005-0323] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Leukemia inhibitory factor (LIF) is required to maintain pluripotency and permit self-renewal of murine embryonic stem (ES) cells. LIF binds to a receptor complex of LIFR-beta and gp130 and signals via the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway, with signalling attenuated by suppressor of cytokine signalling (SOCS) proteins. Recent in vivo studies have highlighted the role of SOCS-3 in the negative regulation of signalling via gp130. To determine the role of SOCS-3 in ES cell biology, SOCS-3-null ES cell lines were generated. When cultured in LIF levels that sustain self-renewal of wild-type cells, SOCS-3-null ES cell lines exhibited less self-renewal and greater differentiation into primitive endoderm. The absence of SOCS-3 enhanced JAK-STAT and extracellular signal-related kinase 1/2 (ERK-1/2)-mitogen-activated protein kinase (MAPK) signal transduction via gp130, with higher levels of phosphorylated STAT-1, STAT-3, SH-2 domain-containing cytoplasmic protein tyrosine phosphatase 2 (SHP-2), and ERK-1/2 in steady state and in response to LIF stimulation. Attenuation of ERK signalling by the addition of MAPK/ERK kinase (MEK) inhibitors to SOCS-3-null ES cell cultures rescued the differentiation phenotype, but did not restore proliferation to wild-type levels. In summary, SOCS-3 plays a crucial role in the regulation of the LIF signalling pathway in murine ES cells. Its absence perturbs the balance between activation of the JAK-STAT and SHP-2-ERK-1/2-MAPK pathways, resulting in less self-renewal and a greater potential for differentiation into the primitive endoderm lineage.
Collapse
Affiliation(s)
- Ariel Forrai
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
464
|
Hoffman LM, Hall L, Batten JL, Young H, Pardasani D, Baetge EE, Lawrence J, Carpenter MK. X-inactivation status varies in human embryonic stem cell lines. Stem Cells 2005; 23:1468-78. [PMID: 16123389 DOI: 10.1634/stemcells.2004-0371] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human embryonic stem cells (hESCs) derived from human blastocysts have an apparently unlimited proliferative capacity and can differentiate into ectoderm, mesoderm, and endoderm. As such, hESC lines have enormous potential for use in cell replacement therapies. It must first be demonstrated, however, that hESCs maintain a stable karyotype and phenotype and that gene expression is appropriately regulated. To date, different hESC lines exhibit similar patterns of expression of markers associated with pluripotent cells. However, the evaluation of epigenetic status of hESC lines has only recently been initiated. One example of epigenetic gene regulation is dosage compensation of the X chromosome in mammalian females. This is achieved through an epigenetic event referred to as X-chromosome inactivation (XCI), an event initiated upon cellular differentiation. We provide the first evidence that undifferentiated hESC lines exhibit different patterns of XCI.
Collapse
Affiliation(s)
- Lisa M Hoffman
- Robarts Research Institute, Krembil Centre for Stem Cell Biology, London, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
465
|
Wiblin AE, Cui W, Clark AJ, Bickmore WA. Distinctive nuclear organisation of centromeres and regions involved in pluripotency in human embryonic stem cells. J Cell Sci 2005; 118:3861-8. [PMID: 16105879 DOI: 10.1242/jcs.02500] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Nuclear organisation is thought to be important in regulating gene expression. Here we investigate whether human embryonic stem cells (hES) have a particular nuclear organisation, which could be important for maintaining their pluripotent state. We found that whereas the nuclei of hES cells have a general gene-density-related radial organisation of chromosomes, as is seen in differentiated cells, there are also distinctive localisations for chromosome regions and gene loci with a role in pluripotency. Chromosome 12p, a region of the human genome that contains clustered pluripotency genes including NANOG, has a more central nuclear localisation in ES cells than in differentiated cells. On chromosome 6p we find no overall change in nuclear chromosome position, but instead we detect a relocalisation of the OCT4 locus, to a position outside its chromosome territory. There is also a smaller proportion of centromeres located close to the nuclear periphery in hES cells compared to differentiated cells. We conclude that hES cell nuclei have a distinct nuclear architecture, especially at loci involved in maintaining pluripotency. Understanding this level of hES cell biology provides a framework within which other large-scale chromatin changes that may accompany differentiation can be considered.
Collapse
Affiliation(s)
- Anne E Wiblin
- MRC Human Genetics Unit, Crewe Road, Edinburgh, EH4 2XU, UK
| | | | | | | |
Collapse
|
466
|
Son YS, Park JH, Kang YK, Park JS, Choi HS, Lim JY, Lee JE, Lee JB, Ko MS, Kim YS, Ko JH, Yoon HS, Lee KW, Seong RH, Moon SY, Ryu CJ, Hong HJ. Heat shock 70-kDa protein 8 isoform 1 is expressed on the surface of human embryonic stem cells and downregulated upon differentiation. Stem Cells 2005; 23:1502-13. [PMID: 16100000 DOI: 10.1634/stemcells.2004-0307] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The cell-surface markers used routinely to define the undifferentiated state and pluripotency of human embryonic stem cells (hESCs) are those used in mouse embryonic stem cells (mESCs) because of a lack of markers directly originated from hESC itself. To identify more hESC-specific cell-surface markers, we generated a panel of monoclonal antibodies (MAbs) by immunizing the irradiated cell clumps of hESC line Miz-hES1, and selected 26 MAbs that were able to bind to Miz-hES1 cells but not to mESCs, mouse embryonic fibroblast cells, and STO cells. Most antibodies did not bind to human neural progenitor cells derived from the Miz-hES1 cells, either. Of these, MAb 20-202S (IgG1, kappa) immunoprecipitated a cell-surface protein of 72-kDa from the lysate of biotin-labeled Miz-hES1 cells, which was identified to be heat shock 70-kDa protein 8 isoform 1 (HSPA8) by quadrupole time-of-flight tandem mass spectrometry. Immunocytochemical analyses proved that the HSPA8 protein was also present on the surface of hESC lines Miz-hES4, Miz-hES6, and HSF6. Two-color flow cytometric analysis of Miz-hES1 and HSF6 showed the coexpression of the HSPA8 protein with other hESC markers such as stage-specific embryonic antigen 3 (SSEA3), SSEA4, TRA-1-60, and TRA-1-81. Flow cytometric and Western blot analyses using various cells showed that MAb 20-202S specifically bound to the HSPA8 protein on the surface of Miz-hES1, contrary to other anti-HSP70 antibodies examined. Furthermore, the surface expression of the HSPA8 protein on Miz-hES1 was markedly downregulated upon differentiation. These data indicate that a novel MAb 20-202S recognizes the HSPA8 protein on the surface of hESCs and suggest that the HSPA8 protein is a putative cell-surface marker for undifferentiated hESCs.
Collapse
Affiliation(s)
- Yeon Sung Son
- Laboratory of Antibody Engineering, Korea Research Institute of Bioscience and Biotechnology, Yusong-Gu, Daejon 305-333, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
467
|
Skottman H, Mikkola M, Lundin K, Olsson C, Strömberg AM, Tuuri T, Otonkoski T, Hovatta O, Lahesmaa R. Gene expression signatures of seven individual human embryonic stem cell lines. Stem Cells 2005; 23:1343-56. [PMID: 16081666 DOI: 10.1634/stemcells.2004-0341] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Identification of molecular components that define a pluripotent human embryonic stem cell (hESC) provides the basis for understanding the molecular mechanisms regulating the maintenance of pluripotency and induction of differentiation. We compared the gene expression profiles of seven genetically independent hESC lines with those of nonlineage-differentiated cells derived from each line. A total of 8,464 transcripts were expressed in all hESC lines. More than 45% of them have no yet-known biological function, which indicates that a high number of unknown factors contribute to hESC pluripotency. Among these 8,464 transcripts, 280 genes were specific for hESCs and 219 genes were more than twofold differentially expressed in all hESC lines compared with nonlineage-differentiated cells. They represent genes implicated in the maintenance of pluripotency and those involved in early differentiation. The chromosomal distribution of these hESC-enriched genes showed over-representation in chromosome 19 and under-representation in chromosome 18. Although the overall gene expression profiles of the seven hESC lines were markedly similar, each line also had a subset of differentially expressed genes reflecting their genetic variation and possibly preferential differentiation potential. Limited overlap between gene expression profiles illustrates the importance of cross-validation of results between different ESC lines.
Collapse
Affiliation(s)
- Heli Skottman
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, POB 123, 0520 Turku, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
468
|
Kang HB, Kim JS, Kwon HJ, Nam KH, Youn HS, Sok DE, Lee Y. Basic Fibroblast Growth Factor Activates ERK and Induces c-Fos in Human Embryonic Stem Cell Line MizhES1. Stem Cells Dev 2005; 14:395-401. [PMID: 16137228 DOI: 10.1089/scd.2005.14.395] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Human embryonic stem (hES) cells can be maintained in a proliferative undifferentiated state in vitro by growing them on feeder layers of mouse embryonic fibroblast (MEF) cells along with basic fibroblast growth factor (bFGF/FGF-2). To understand the molecular mechanisms involved in the requirement of bFGF in human ES cells, we investigated expression of FGF receptors and intracellular signaling events in response to bFGF in human ES cell line MizhES1. On the basis of the results of RT-PCR, clear expression of FGF receptors FGFR1, FGR2, and FGFR3 was noticed. Because MAPK, PI3K, and PKC pathways are well-known pathways triggered by bFGF in other cells, these pathways were investigated after stimulation with bFGF. bFGF did not induce activation of PI3K or PKC, but induced activation of ERK (extracellular signal-regulated kinase). To monitor the consequences of ERK activation, we examined expression of the immediate early gene c-fos, one downstream target of the MEK1/ERK pathway. mRNA and protein levels of the c-fos gene were increased by bFGF. Induction of c-Fos was dependent on MEKl. Therefore, it is likely that bFGF contributes to maintenance of human ES cells, at least in part, through the MEK1/ERK pathway.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Line
- Cells, Cultured
- Collagen/pharmacology
- Culture Media, Serum-Free/pharmacology
- DNA-Binding Proteins/metabolism
- Drug Combinations
- Enzyme Activation
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Fibroblast Growth Factor 2/metabolism
- Fluorescent Antibody Technique, Indirect
- Humans
- Immunoblotting
- Laminin/pharmacology
- Mice
- Microscopy, Confocal
- Octamer Transcription Factor-3
- Organic Cation Transport Proteins/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Kinase C/metabolism
- Protein-Tyrosine Kinases/metabolism
- Proteoglycans/pharmacology
- Proto-Oncogene Proteins c-fos/metabolism
- RNA, Messenger/metabolism
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptor, Fibroblast Growth Factor, Type 2
- Receptor, Fibroblast Growth Factor, Type 3
- Receptors, Fibroblast Growth Factor/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Stem Cells/cytology
- Time Factors
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Ho Bum Kang
- Department of Biochemistry, Biotechnology Research Institute, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 361-763, Korea
| | | | | | | | | | | | | |
Collapse
|
469
|
Abstract
Stem cells are unique in their capacity to self-renew and generate differentiated progeny to maintain tissues throughout life. A common molecular program for stem cells has remained elusive. We discuss what the molecular logic of stemness may be. We suggest that it may not be coupled to distinct cellular properties such as self-renewal or multipotency, but rather to the stable suspension at a specific developmental stage. In this view, the stem cell niche allows a cell to maintain a transcriptional accessibility enabling the generation of specific differentiated progeny.
Collapse
Affiliation(s)
- Harald Mikkers
- Department of Cell and Molecular Biology, Medical Nobel Institute, Karolinska Institute, Stockholm, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Medical Nobel Institute, Karolinska Institute, Stockholm, Sweden
- Department of Cell and Molecular Biology, Medical Nobel Institute, Karolinska Institute, Box 285, 17177 Stockholm, Sweden. Tel.: +46 8 524 87562; Fax: +46 8 324927; E-mail:
| |
Collapse
|
470
|
Prindull G. Hypothesis: Cell plasticity, linking embryonal stem cells to adult stem cell reservoirs and metastatic cancer cells? Exp Hematol 2005; 33:738-46. [PMID: 15963849 DOI: 10.1016/j.exphem.2005.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Revised: 02/24/2005] [Accepted: 03/02/2005] [Indexed: 11/26/2022]
Abstract
Embryonal stem (ES) cells are the earliest ontogenetically identifiable stem cells of the embryo proper for all subsequent mesenchymal stem cells and for highly specialized differentiated cells. This review characterizes, in a working hypothesis, the role of reversible EMT/MET (epithelialmesenchymal transition) as a manifestation of cell plasticity 1) in the development of ES cells to adult stem cells (hematopoietic stem cells) and 2) in metastasizing cancer cells. Animal studies support the concept that EMT/MET is a key manifestation of cell plasticity in the development of ES cells to adult stem cells, and in conversion of localized to metastasizing cancer cells. In fact, ES cells may persist to postnatal life, in cytologically verifiable form and/or within the frame of EMT/MET, as ultimate reservoir for adult stem cells. Furthermore, EMT could possibly serve as a conceptional link between physiologic and pathologic signaling pathways. Clonal confirmation in humans is necessary.
Collapse
Affiliation(s)
- Gregor Prindull
- Department of Pediatrics, University of Göttingen Medical School, Germany.
| |
Collapse
|
471
|
Boer B, Luster TA, Bernadt C, Rizzino A. Distal enhancer of the mouseFGF-4 gene and its human counterpart exhibit differential activity: Critical role of a GT box. Mol Reprod Dev 2005; 71:263-74. [PMID: 15803454 DOI: 10.1002/mrd.20264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Previous studies have shown that there is a strict requirement for fibroblast growth factor-4 (FGF-4) during mammalian embryogenesis, and that FGF-4 expression in embryonic stem (ES) cells and embryonal carcinoma (EC) cells are controlled by a powerful downstream distal enhancer. More recently, mouse ES cells were shown to express significantly more FGF-4 mRNA than human ES cells. In the work reported here, we demonstrate that mouse EC cells also express far more FGF-4 mRNA than human EC cells. Using a panel of FGF-4 promoter/reporter gene constructs, we demonstrate that the enhancer of the mouse FGF-4 gene is approximately tenfold more active than its human counterpart. Moreover, we demonstrate that the critical difference between the mouse and the human FGF-4 enhancer is a 4 bp difference in the sequence of an essential GT box. Importantly, we demonstrate that changing 4 bp in the human enhancer to match the sequence of the mouse GT box elevates the activity of the human FGF-4 enhancer to the same level as that of the mouse enhancer. We extended these studies by examining the roles of Sp1 and Sp3 in FGF-4 expression. Although we demonstrate that Sp3, but not Sp1, can activate the FGF-4 promoter when artificially tethered to the FGF-4 enhancer, we show that Sp3 is not essential for expression of FGF-4 mRNA in mouse ES cells. Finally, our studies with human EC cells suggest that the factor responsible for mediating the effect of the mouse GT box is unlikely to be Sp1 or Sp3, and this factor is either not expressed in human EC cells or it is not sufficiently active in these cells.
Collapse
Affiliation(s)
- Brian Boer
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-6805, USA
| | | | | | | |
Collapse
|
472
|
Abstract
The discovery of mouse embryonic stem (ES) cells >20 years ago represented a major advance in biology and experimental medicine, as it enabled the routine manipulation of the mouse genome. Along with the capacity to induce genetic modifications, ES cells provided the basis for establishing an in vitro model of early mammalian development and represented a putative new source of differentiated cell types for cell replacement therapy. While ES cells have been used extensively for creating mouse mutants for more than a decade, their application as a model for developmental biology has been limited and their use in cell replacement therapy remains a goal for many in the field. Recent advances in our understanding of ES cell differentiation, detailed in this review, have provided new insights essential for establishing ES cell-based developmental models and for the generation of clinically relevant populations for cell therapy.
Collapse
Affiliation(s)
- Gordon Keller
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA.
| |
Collapse
|
473
|
Dvorak P, Dvorakova D, Koskova S, Vodinska M, Najvirtova M, Krekac D, Hampl A. Expression and potential role of fibroblast growth factor 2 and its receptors in human embryonic stem cells. Stem Cells 2005; 23:1200-11. [PMID: 15955829 DOI: 10.1634/stemcells.2004-0303] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although the detection of several components of the fibroblast growth factor (FGF) signaling pathway in human embryonic stem cells (hESCs) has been reported, the functionality of that pathway and effects on cell fate decisions are yet to be established. In this study we characterized expression of FGF-2, the prototypic member of the FGF family, and its receptors (FGFRs) in undifferentiated and differentiating hESCs; subsequently, we analyzed the effects of FGF-2 on hESCs, acting as both exogenous and endogenous factors. We have determined that undifferentiated hESCs are abundant in several molecular-mass isoforms of FGF-2 and that expression pattern of these isoforms remains unchanged under conditions that induce hESC differentiation. Significantly, FGF-2 is released by hESCs into the medium, suggesting an autocrine activity. Expression of FGFRs in undifferentiated hESCs follows a specific pattern, with FGFR1 being the most abundant species and other receptors showing lower expression in the following order: FGFR1 --> FGFR3 --> FGFR4 --> FGFR2. Initiation of differentiation is accompanied by profound changes in FGFR expression, particularly the upregulation of FGFR1. When hESCs are exposed to exogenous FGF-2, extracellular signal-regulated kinases are phosphorylated and thereby activated. However, the presence or absence of exogenous FGF-2 does not significantly affect the proliferation of hESCs. Instead, increased concentration of exogenous FGF-2 leads to reduced outgrowth of hESC colonies with time in culture. Finally, the inhibitor of FGFRs, SU5402, was used to ascertain whether FGF-2 that is released by hESCs exerts its activities via autocrine pathways. Strikingly, the resultant inhibition of FGFR suppresses activation of downstream protein kinases and causes rapid cell differentiation, suggesting an involvement of autocrine FGF signals in the maintenance of proliferating hESCs in the undifferentiated state. In conclusion from our data, we propose that this endogenous FGF signaling pathway can be implicated in self-renewal or differentiation of hESCs.
Collapse
Affiliation(s)
- Petr Dvorak
- Laboratory of Molecular Embryology, Mendel University, Brno Zemedelska 1, 613 00 Brno, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
474
|
Abstract
Human embryonic stem cells have been defined as self-renewing cells that can give rise to many types of cells of the body. How and whether these cells can be manipulated to replace cells in diseased tissues, used to screen drugs and toxins, or studied to better understand normal development, however, depends on knowing more about their fundamental properties. Many different human embryonic stem cell lines--which are pluripotent, proliferate indefinitely in vitro and maintain a normal, euploid karyotype over extended culture--have now been derived, but whether these cell lines are in fact equivalent remains unclear. It will therefore be important to define robust criteria for the assessment of both existing and newly derived cell lines and for the validation of new culture conditions.
Collapse
Affiliation(s)
- Lisa M Hoffman
- Stem Cell Biology and Regenerative Medicine, Robarts Research Institute, 100 Perth Drive, London, Ontario N6A 5K8, Canada
| | | |
Collapse
|
475
|
Miura T, Luo Y, Khrebtukova I, Brandenberger R, Zhou D, Thies RS, Vasicek T, Young H, Lebkowski J, Carpenter MK, Rao MS. Monitoring early differentiation events in human embryonic stem cells by massively parallel signature sequencing and expressed sequence tag scan. Stem Cells Dev 2005; 13:694-715. [PMID: 15684837 DOI: 10.1089/scd.2004.13.694] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
To identify genes that may be involved in the process of human embryonic stem cell (hESC) differentiation, we profiled gene expression by expressed sequenced tag (EST) enumeration and massively parallel signature sequencing (MPSS) using RNA samples from feeder-free cultures of undifferentiated (passages 40-50) and differentiated (day 14) H1, H7, and H9 lines. MPSS and EST scan analysis showed good concordance and identified a large number of genes that changed rapidly as cultures transition from a pluripotent to a differentiated state. These included known and unknown ES cell-specific genes as well as a large number of known genes that were altered as cells differentiate. A subset of genes that were either up- or down-regulated were selected and their differential expression confirmed by a variety of independent methods, including comparison of expression after further differentiation, publicly available databases, and direct assessments by reverse transcriptase (RT)-PCR and immunocytochemistry. The analysis identified markers unique to the hESC and embryoid bodies (hEBs) stage as well as signaling pathways that likely regulate differentiation. The data generated can be used to monitor the state of hESC isolated by different laboratories using independent methods and maintained under differing culture conditions.
Collapse
Affiliation(s)
- Takumi Miura
- Gerontology Research Center, Stem Cell Biology Unit/Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
476
|
Brimble SN, Zeng X, Weiler DA, Luo Y, Liu Y, Lyons IG, Freed WJ, Robins AJ, Rao MS, Schulz TC. Karyotypic stability, genotyping, differentiation, feeder-free maintenance, and gene expression sampling in three human embryonic stem cell lines derived prior to August 9, 2001. Stem Cells Dev 2005; 13:585-97. [PMID: 15684826 DOI: 10.1089/scd.2004.13.585] [Citation(s) in RCA: 164] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The number of human embryonic stem cell (hESC) lines available to federally funded U.S. researchers is currently limited. Thus, determining their basic characteristics and disseminating these lines is important. In this report, we recovered and expanded the earliest available cryopreserved stocks of the BG01, BG02, and BG03 hESC lines. These cultures exhibited multiple definitive characteristics of undifferentiated cells, including long-term self-renewal, expression of markers of pluripotency, maintenance of a normal karyotype, and differentiation to mesoderm, endoderm, and ectoderm. Each cell line exhibited a unique genotype and human leukocyte antigen (HLA) isotype, confirming that they were isolated independently. BG01, BG02, and BG03 maintained in feederfree conditions demonstrated self-renewal, maintenance of normal karyotype, and gene expression indicative of undifferentiated pluripotent stem cells. A survey of gene expression in BG02 cells using massively parallel signature sequencing generated a digital read-out of transcript abundance and showed that this line was similar to other hESC lines. BG01, BG02, and BG03 hESCs are therefore independent, undifferentiated, and pluripotent lines that can be maintained without accumulation of karyotypic abnormalities.
Collapse
|
477
|
McConnell J, Petrie L, Stennard F, Ryan K, Nichols J. Eomesodermin
is expressed in mouse oocytes and pre-implantation embryos. Mol Reprod Dev 2005; 71:399-404. [PMID: 15880683 DOI: 10.1002/mrd.20318] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
T-box genes are a highly conserved family of genes encoding transcription factors, which share a conserved DNA binding domain (the T-box). Appropriate temporal and spatial expression of this gene family is critical for gastrulation and organogenesis in a number of species. The T-box containing gene Eomesodermin was first identified in Xenopus, where it plays a critical role in mesoderm formation. In situ analyses in mice have described the expression patterns of the mouse ortholog of this gene mEomesodermin (mEomes) at the time of implantation and during fetal development. Additional studies involving the disruption of the mEomes gene, have demonstrated an additional role for mEomes in trophoblast formation. However, these analyses did not address the possibility that maternally encoded or pre-blastocyst zygotic transcription of mEomes may also contribute to embryonic development. We show here that mEomes mRNA is present prior to blastocyst formation, and that the protein product of mEomes is associated with nuclear DNA during oocyte development and persistently localizes within all nuclei of the preimplantation embryo until the early blastocyst stage. mEomes protein is associated with the meiotic spindle in the unfertilized egg and with the mitotic spindle at each cell division. Our results are consistent with mEomesodermin having a role in early preimplantation development and inner cell mass formation in addition to its function in the trophoblast lineage.
Collapse
Affiliation(s)
- Josie McConnell
- Rowett Research Institute, Bucksburn, Aberdeen, United Kingdom.
| | | | | | | | | |
Collapse
|
478
|
Abstract
Several recent reports used microarray, serial analysis of gene expression (SAGE), and expressed sequence tags (EST) strategies to characterize the human embryonic stem cell transcriptome and those of their differentiated derivatives. All three approaches yielded valuable data and highlight the fact that a large percentage of genes remain uncharacterized in these cells representative of early human development.
Collapse
Affiliation(s)
- Paul Robson
- Stem Cell and Developmental Biology, Genome Institute of Singapore and Department of Biological Sciences, National University of Singapore, 60 Biopolis Street, 02-01, Genome Building, Singapore 138672.
| |
Collapse
|
479
|
Palmqvist L, Glover CH, Hsu L, Lu M, Bossen B, Piret JM, Humphries RK, Helgason CD. Correlation of Murine Embryonic Stem Cell Gene Expression Profiles with Functional Measures of Pluripotency. Stem Cells 2005; 23:663-80. [PMID: 15849174 DOI: 10.1634/stemcells.2004-0157] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Global gene expression profiling was performed on murine embryonic stem cells (ESCs) induced to differentiate by removal of leukemia inhibitory factor (LIF) to identify genes whose change in expression correlates with loss of pluripotency. To identify appropriate time points for the gene expression analysis, the dynamics of loss of pluripotency were investigated using three functional assays: chimeric mouse formation, embryoid body generation, and colony-forming ability. A rapid loss of pluripotency was detected within 24 hours, with very low residual activity in all assays by 72 hours. Gene expression profiles of undifferentiated ESCs and ESCs cultured for 18 and 72 hours in the absence of LIF were determined using the Affymetrix GeneChip U74v2. In total, 473 genes were identified as significantly differentially expressed, with approximately one third having unknown biological function. Among the 275 genes whose expression decreased with ESC differentiation were several factors previously identified as important for, or markers of, ESC pluripotency, including Stat3, Rex1, Sox2, Gbx2, and Bmp4. A significant number of the decreased genes also overlap with previously published mouse and human ESC data. Furthermore, several membrane proteins were among the 48 decreased genes correlating most closely with the functional assays, including the stem cell factor receptor c-Kit. Through identification of genes whose expression closely follows functional properties of ESCs during early differentiation, this study lays the foundation for further elucidating the molecular mechanisms regulating the maintenance of ESC pluripotency and facilitates the identification of more reliable molecular markers of the undifferentiated state.
Collapse
Affiliation(s)
- Lars Palmqvist
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
480
|
Abstract
FGF2 is a key regulator of survival and proliferation of mammalian CNS stem cells. Cells within undifferentiated rodent neurospheres express FGF receptors (FGFRs), but their expression patterns and potential roles in human neurosphere proliferation and differentiation have not been examined. Our aim was to provide an initial overview of the relative profiles of FGFRs before and after differentiation of human neurospheres derived either from embryonic brain or spinal cord. In 'undifferentiated' neurospheres, transcripts from FGFR1 and FGFR2 were consistently detected. FGFR3 could be detected in undifferentiated brain neurospheres and in spinal cord early neurospheres. Following differentiation the most dramatic and consistent change was a decrease in FGFR1 mRNA, suggesting a role for this receptor in maintenance of the undifferentiated state.
Collapse
Affiliation(s)
- Melanie Mackay
- Developmental Biology Unit, Institute of Child Health, UCL, 30 Guilford Street, London WC1N 1EH, UK
| | | |
Collapse
|
481
|
Wobus AM, Boheler KR. Embryonic stem cells: prospects for developmental biology and cell therapy. Physiol Rev 2005; 85:635-78. [PMID: 15788707 DOI: 10.1152/physrev.00054.2003] [Citation(s) in RCA: 461] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Stem cells represent natural units of embryonic development and tissue regeneration. Embryonic stem (ES) cells, in particular, possess a nearly unlimited self-renewal capacity and developmental potential to differentiate into virtually any cell type of an organism. Mouse ES cells, which are established as permanent cell lines from early embryos, can be regarded as a versatile biological system that has led to major advances in cell and developmental biology. Human ES cell lines, which have recently been derived, may additionally serve as an unlimited source of cells for regenerative medicine. Before therapeutic applications can be realized, important problems must be resolved. Ethical issues surround the derivation of human ES cells from in vitro fertilized blastocysts. Current techniques for directed differentiation into somatic cell populations remain inefficient and yield heterogeneous cell populations. Transplanted ES cell progeny may not function normally in organs, might retain tumorigenic potential, and could be rejected immunologically. The number of human ES cell lines available for research may also be insufficient to adequately determine their therapeutic potential. Recent molecular and cellular advances with mouse ES cells, however, portend the successful use of these cells in therapeutics. This review therefore focuses both on mouse and human ES cells with respect to in vitro propagation and differentiation as well as their use in basic cell and developmental biology and toxicology and presents prospects for human ES cells in tissue regeneration and transplantation.
Collapse
Affiliation(s)
- Anna M Wobus
- In Vitro Differentiation Group, IPK Gatersleben, Germany.
| | | |
Collapse
|
482
|
Jamal AM, Lipsett M, Sladek R, Laganière S, Hanley S, Rosenberg L. Morphogenetic plasticity of adult human pancreatic islets of Langerhans. Cell Death Differ 2005; 12:702-12. [PMID: 15818398 DOI: 10.1038/sj.cdd.4401617] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to investigate the phenotypic plasticity of pancreatic islets of Langerhans. Quiescent adult human islets were induced to undergo a phenotypic switch to highly proliferative duct-like structures in a process characterized by a loss of expression of islet-specific hormones and transcription factors as well as a temporally related rise in the expression of markers of both duct epithelial and progenitor cells. Short-term treatment of these primitive duct-like structures with the neogenic factor islet neogenesis-associated protein (INGAP104-118) induced their reconversion back to islet-like structures in a PI3-kinase-dependent manner. These neoislets resembled freshly isolated human islets with respect to the presence and topological arrangement of the four endocrine cell types, islet gene expression and hormone production, insulin content and glucose-responsive insulin secretion. Our results suggest that adult human islets possess a remarkable degree of morphogenetic plasticity. This novel observation may have important implications for understanding pancreatic carcinogenesis and islet neogenesis.
Collapse
Affiliation(s)
- A-M Jamal
- Department of Surgery, McGill University, Montréal, Canada
| | | | | | | | | | | |
Collapse
|
483
|
Zeng X, Cai J, Chen J, Luo Y, You ZB, Fotter E, Wang Y, Harvey B, Miura T, Backman C, Chen GJ, Rao MS, Freed WJ. Dopaminergic differentiation of human embryonic stem cells. ACTA ACUST UNITED AC 2005; 22:925-40. [PMID: 15536184 DOI: 10.1634/stemcells.22-6-925] [Citation(s) in RCA: 291] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this manuscript we report that human embryonic stem cells (hESCs) differentiated into dopaminergic neurons when cocultured with PA6 cells. After 3 weeks of differentiation, approximately 87% of hES colonies contained tyrosine hydroxylase (TH)-positive cells, and a high percentage of the cells in most of the colonies expressed TH. Differentiation was inhibited by exposure to BMP4 or serum. TH-positive cells derived from hESCs were postmitotic, as determined by bromodeoxyurindine colabeling. Differentiated cells expressed other markers of dopaminergic neurons, including the dopamine transporter, aromatic amino acid decarboxylase, and the transcription factors associated with neuronal and dopaminergic differentiation, Sox1, Nurr1, Ptx3, and Lmx1b. Neurons that had been differentiated on PA6 cells were negative for dopamine-beta-hydroxylase, a marker of noradrenergic neurons. PA6-induced neurons were able to release dopamine and 3,4-dihydroxphe-hylacetic acid (DOPAC) but not noradrenalin when depolarized by high K(+). When transplanted into 6-hydroxydopamine-treated animals, hES-derived dopaminergic cells integrated into the rat striatum. Five weeks after transplantation, surviving TH-positive cells were present but in very small numbers compared with the high frequency of TH-positive cells seen in PA6 coculture. Larger numbers of cells positive for smooth muscle actin, but no undifferentiated ES cells, were present after transplantation. Therefore, hESCs can be used to generate human dopaminergic cells that exhibit biochemical and functional properties consistent with the expected properties of mature dopaminergic neurons.
Collapse
Affiliation(s)
- Xianmin Zeng
- Cellular Neurobiology Research Branch, National Institute on Drug Abuse, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
484
|
Sonntag KC, Simantov R, Isacson O. Stem cells may reshape the prospect of Parkinson's disease therapy. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2005; 134:34-51. [PMID: 15790528 DOI: 10.1016/j.molbrainres.2004.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/09/2004] [Indexed: 12/21/2022]
Abstract
The concept of cell replacement to compensate for cell loss and restore functionality has entered several disease entities including neurodegenerative disorders. Recent clinical studies have shown that transplantation of fetal dopaminergic (DA) cells into the brain of Parkinson's disease (PD) patients can reduce disease-associated motor deficits. However, the use of fetal tissue is associated with practical and ethical problems including low efficiency, variability in the clinical outcome and controversy regarding the use of fetuses as donor. An alternative cell resource could be embryonic stem (ES) cells, which can be cultivated in unlimited amounts and which have the potential to differentiate into mature DA cells. Several differentiation protocols have been developed, and some progress has been made in understanding the mechanisms underlying DA specification in ES cell development, but the "holy grail" in this paradigm, which is the production of sufficient amounts of the "right" therapeutic DA cell, has not yet been accomplished. To achieve this goal, several criteria on the transplanted DA cells need to be fulfilled, mainly addressing cell survival, accurate integration in the brain circuitry, normal function, no tumor formation, and no immunogenicity. Here, we summarize the current state of ES cell-derived DA neurogenesis and discuss the aspects involved in generating an optimal cell source for cell replacement in PD.
Collapse
Affiliation(s)
- Kai-Christian Sonntag
- Udall Parkinson's Disease Research Center of Excellence, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478, USA.
| | | | | |
Collapse
|
485
|
Wang L, Duan E, Sung LY, Jeong BS, Yang X, Tian XC. Generation and characterization of pluripotent stem cells from cloned bovine embryos. Biol Reprod 2005; 73:149-55. [PMID: 15744021 DOI: 10.1095/biolreprod.104.037150] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bovine embryonic stem (ES) cell lines reported to date vary in morphology and marker expression (e.g., alkaline phosphatase [ALPL], stage-specific embryonic antigen 4 [SSEA4], and OCT4) that normally are associated with the undifferentiated, pluripotent state. These observations suggest that the proper experimental conditions for consistently producing bovine ES cells have not been identified. Here, we report three bovine ES cell lines, one from in vitro-fertilized and two from nuclear transfer embryos. These bovine ES cells grew in large, multicellular colonies resembling the mouse ES and embryonic germ (EG) cells and human EG cells. Throughout the culture period, most of the cells within the colonies stained positive for ALPL and the cell surface markers SSEA4 and OCT4. The staining patterns of nuclear transfer ES cells were identical to those of the blastocysts generated in vitro yet different from most previously reported bovine ES cell lines, which were either negative or not detected. After undifferentiated culture for more than 1 yr, these cells maintained the ability to differentiate into embryoid bodies and derivatives of all three EG layers, thus demonstrating their pluripotency. However, unlike the mouse and human ES cells, following treatment with trypsin, type IV collagenase, or protease E, our bovine ES cells failed to self-renew and became spontaneously differentiated. Presumably, this resulted from an interruption of the self-renewal pathway. In summary, we generated pluripotent bovine ES cells with morphology similar to those of established ES cells in humans and mice as well as marker-staining patterns identical to those of the bovine blastocysts.
Collapse
Affiliation(s)
- Li Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing
| | | | | | | | | | | |
Collapse
|
486
|
Li XJ, Du ZW, Zarnowska ED, Pankratz M, Hansen LO, Pearce RA, Zhang SC. Specification of motoneurons from human embryonic stem cells. Nat Biotechnol 2005; 23:215-21. [PMID: 15685164 DOI: 10.1038/nbt1063] [Citation(s) in RCA: 568] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2004] [Accepted: 11/04/2004] [Indexed: 02/07/2023]
Abstract
An understanding of how mammalian stem cells produce specific neuronal subtypes remains elusive. Here we show that human embryonic stem cells generated early neuroectodermal cells, which organized into rosettes and expressed Pax6 but not Sox1, and then late neuroectodermal cells, which formed neural tube-like structures and expressed both Pax6 and Sox1. Only the early, but not the late, neuroectodermal cells were efficiently posteriorized by retinoic acid and, in the presence of sonic hedgehog, differentiated into spinal motoneurons. The in vitro-generated motoneurons expressed HB9, HoxC8, choline acetyltransferase and vesicular acetylcholine transporter, induced clustering of acetylcholine receptors in myotubes, and were electrophysiologically active. These findings indicate that retinoic acid action is required during neuroectoderm induction for motoneuron specification and suggest that stem cells have restricted capacity to generate region-specific projection neurons even at an early developmental stage.
Collapse
|
487
|
Vallier L, Reynolds D, Pedersen RA. Nodal inhibits differentiation of human embryonic stem cells along the neuroectodermal default pathway. Dev Biol 2005; 275:403-21. [PMID: 15501227 DOI: 10.1016/j.ydbio.2004.08.031] [Citation(s) in RCA: 282] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2004] [Revised: 08/12/2004] [Accepted: 08/20/2004] [Indexed: 01/04/2023]
Abstract
Genetic studies in fish, amphibia, and mice have shown that deficiency of Nodal signaling blocks differentiation into mesoderm and endoderm. Thus, Nodal is considered as a major inducer of mesendoderm during gastrulation. On this basis, Nodal is a candidate for controlling differentiation of pluripotent human embryonic stem cells (hESCs) into tissue lineages with potential clinical value. We have investigated the effect of Nodal, both as a recombinant protein and as a constitutively expressed transgene, on differentiation of hESCs. When control hESCs were grown in chemically defined medium, their expression of markers of pluripotency progressively decreased, while expression of neuroectoderm markers was strongly upregulated, thus revealing a neuroectodermal default mechanism for differentiation in this system. hESCs cultured in recombinant Nodal, by contrast, showed prolonged expression of pluripotency marker genes and reduced induction of neuroectoderm markers. These Nodal effects were accentuated in hESCs expressing a Nodal transgene, with striking morphogenetic consequences. Nodal-expressing hESCs developing as embryoid bodies contained an outer layer of visceral endoderm-like cells surrounding an inner layer of epiblast-like cells, each layer having distinct gene expression patterns. Markers of neuroectoderm were not upregulated during development of Nodal-expressing embryoid bodies, nor was there induction of markers for definitive mesoderm or endoderm differentiation. Moreover, the inner layer expressed markers of pluripotency, characteristic of undifferentiated hESCs and of epiblast in mouse embryos. These results could be accounted for by an inhibitory effect of Nodal-induced visceral endoderm on pluripotent cell differentiation into mesoderm and endoderm, with a concomitant inhibition of neuroectoderm differentiation by Nodal itself. There could also be a direct effect of Nodal in the maintenance of pluripotency. In summary, analysis of the Nodal-expressing phenotype suggests a function for the transforming growth factor-beta (TGF-beta) growth factor superfamily in pluripotency and in early cell fate decisions leading to primary tissue layers during in vitro development of pluripotent human stem cells. The effects of Nodal on early differentiation illustrate how hESCs can augment mouse embryos as a model for analyzing mechanisms of early mammalian development.
Collapse
Affiliation(s)
- Ludovic Vallier
- Department of Surgery, University of Cambridge, Cambridge CB2 2QQ, United Kingdom.
| | | | | |
Collapse
|
488
|
Abstract
It is widely anticipated that human embryonic stem (ES) cells will serve as an experimental model for studying early development in our species, and, conversely, that studies of development in model systems, the mouse in particular, will inform our efforts to manipulate human stem cells in vitro. A comparison of primate and mouse ES cells suggests that a common underlying blueprint for the pluripotent state has undergone significant species-specific modification. As we discuss here, technical advances in the propagation and manipulation of human ES cells have improved our understanding of their growth and differentiation, providing the potential to investigate early human development and to develop new clinical therapies.
Collapse
Affiliation(s)
- Martin F Pera
- Monash Institute of Reproduction and Development, Monash University, Clayton, Victoria, Australia.
| | | |
Collapse
|
489
|
van de Stolpe A, van den Brink S, van Rooijen M, Ward-van Oostwaard D, van Inzen W, Slaper-Cortenbach I, Fauser B, van den Hout N, Weima S, Passier R, Smith N, Denning C, Mummery C. Human embryonic stem cells: towards therapies for cardiac disease. Derivation of a Dutch human embryonic stem cell line. Reprod Biomed Online 2005; 11:476-85. [PMID: 16274613 DOI: 10.1016/s1472-6483(10)61144-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cell transplantation is being discussed as a potential therapy for multiple disorders caused by loss or malfunction of single or at most a few cell types. These include diabetes, Parkinson's disease and myocardial infarction or cardiac failure. However, it is not yet clear whether cells from adult tissues ('adult stem cells') or embryos ('embryonic stem cells') will prove to be the most appropriate replacement cells; most likely, each disease will have its own preferred source. This study presents the background to this discussion and the current state of research in replacement of cardiac tissue, with focus on recent developments using human embryonic stem cells. It also describes a new human embryonic stem cell (HESC) line, NL-HESC1, the first to be derived in the Netherlands, and shows that it forms cardiac cells in a manner comparable with that of hES2 and hES3 cells grown in the same laboratory.
Collapse
|
490
|
Gjørret JO, Maddox-Hyttel P. Attempts towards derivation and establishment of bovine embryonic stem cell-like cultures. Reprod Fertil Dev 2005; 17:113-24. [PMID: 15745636 DOI: 10.1071/rd04117] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2004] [Accepted: 10/01/2004] [Indexed: 11/23/2022] Open
Abstract
Current knowledge on the biology of mammalian embryonic stem cells (ESC) is stunningly sparse in light of their potential value in studies of development, functional genomics, generation of transgenic animals and human medicine. Despite many efforts to derive ESC from other mammalian species, ESC that retain their capacity for germ line transmission have only been verified in the mouse. However, the criterion of germ line transmission may not need to be fulfilled for exploitation of other abilities of these cells. Promising results with human ESC-like cells and adult stem cells have nourished great expectations for their potential use in regenerative medicine. However, such an application is far from reality and substantial research is required to elucidate aspects of the basic biology of pluripotent cells, as well as safety issues associated with the use of such cells in therapy. In this context, methods for the derivation, propagation and differentiation of ESC-like cultures from domestic animals would be highly desirable as biologically relevant models. Here, we review previously published efforts to establish bovine ESC-like cells and describe a procedure used in attempts to derive similar cells from bovine Day 12 embryos.
Collapse
Affiliation(s)
- Jakob O Gjørret
- Department of Animal and Veterinary Sciences, Royal Veterinary and Agricultural University, Denmark.
| | | |
Collapse
|
491
|
Schulz TC, Noggle SA, Palmarini GM, Weiler DA, Lyons IG, Pensa KA, Meedeniya ACB, Davidson BP, Lambert NA, Condie BG. Differentiation of Human Embryonic Stem Cells to Dopaminergic Neurons in Serum-Free Suspension Culture. Stem Cells 2004; 22:1218-38. [PMID: 15579641 DOI: 10.1634/stemcells.2004-0114] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The use of human embryonic stem cells (hESCs) as a source of dopaminergic neurons for Parkinson's disease cell therapy will require the development of simple and reliable cell differentiation protocols. The use of cell cocultures, added extracellular signaling factors, or transgenic approaches to drive hESC differentiation could lead to additional regulatory as well as cell production delays for these therapies. Because the neuronal cell lineage seems to require limited or no signaling for its formation, we tested the ability of hESCs to differentiate to form dopamine-producing neurons in a simple serum-free suspension culture system. BG01 and BG03 hESCs were differentiated as suspension aggregates, and neural progenitors and neurons were detectable after 2-4 weeks. Plated neurons responded appropriately to electrophysiological cues. This differentiation was inhibited by early exposure to bone morphogenic protein (BMP)-4, but a pulse of BMP-4 from days 5 to 9 caused induction of peripheral neuronal differentiation. Real-time polymerase chain reaction and whole-mount immunocytochemistry demonstrated the expression of multiple markers of the midbrain dopaminergic phenotype in serum-free differentiations. Neurons expressing tyrosine hydroxylase (TH) were killed by 6-hydroxydopamine (6-OHDA), a neurotoxic catecholamine. Upon plating, these cells released dopamine and other catecholamines in response to K+ depolarization. Surviving TH+ neurons, derived from the cells differentiated in serum-free suspension cultures, were detected 8 weeks after transplantation into 6-OHDA-lesioned rat brains. This work suggests that hESCs can differentiate in simple serum-free suspension cultures to produce the large number of cells required for transplantation studies.
Collapse
Affiliation(s)
- Thomas C Schulz
- BresaGen Inc., 111 Riverbend Rd., Athens, Georgia, 30605, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
492
|
Abstract
Most mammalian cells when placed in culture will undergo a limited number of cell divisions before entering an unresponsive non-proliferating state termed senescence. However, several pathways that are activated singly or in concert can allow cells to bypass senescence at least for limited periods. These include the telomerase pathway required to maintain telomere ends, the p53 and Rb pathways required to direct senescence in response to DNA damage, telomere shortening and mitogenic signals, and the insulin-like growth factor--Akt pathway that may regulate lifespan and cell proliferation. In this review, we summarize recent findings related to these pathways in embryonic stem (ES) cells and suggest that ES cells are immortal because these pathways are tightly regulated.
Collapse
Affiliation(s)
- Takumi Miura
- Stem Cell Biology Unit, Laboratory of Neurosciences, National Institute on Aging, Gerontology Research Center, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
493
|
De Sousa PA, da Silva SJM, Anderson RA. Neurotrophin Signaling in Oocyte Survival and Developmental Competence: A Paradigm for Cellular Toti-Potency. CLONING AND STEM CELLS 2004; 6:375-85. [PMID: 15671666 DOI: 10.1089/clo.2004.6.375] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
While not fulfilling the criterion of a "stem cell" in being capable of self-renewal, mature and fertilized oocytes are the original "toti-potent" cells, whose capacity for expansion and differentiation can only be approximated by stem cells of embryonic or adult origin in vitro. As such, the mechanisms by which oocytes acquire and manifest competence to support embryo development is of fundamental interest to efforts to control and re-specify somatic cell fate and toti-potency. This is underscored by the unparalleled capacity of oocyte cytoplasm to successfully re-specify the genetic program of animal development following cell nuclear replacement (i.e., cloning). Thus, the knowledge gained by understanding the acquisition of oocyte developmental competence could ultimately facilitate the creation of adult stem cells in vitro from terminally differentiated cells, ex ovo. In this paper, we review the concept of oocyte developmental competence, and focus on our own research and that of others implicating a role for neurotrophins in this process, and that of oocyte cell survival. Lastly we propose a role for neurotrophin signalling in embryo stem cell survival.
Collapse
Affiliation(s)
- P A De Sousa
- Division of Gene Function and Development, Roslin Institute, Roslin, Midlothian, EH25 9PS, UK.
| | | | | |
Collapse
|
494
|
Rao M. Conserved and divergent paths that regulate self-renewal in mouse and human embryonic stem cells. Dev Biol 2004; 275:269-86. [PMID: 15501218 DOI: 10.1016/j.ydbio.2004.08.013] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2004] [Revised: 08/10/2004] [Accepted: 08/10/2004] [Indexed: 01/10/2023]
Abstract
The past few years have seen remarkable progress in our understanding of embryonic stem cell (ES cell) biology. The necessity of examining human ES cells in culture, coupled with the wealth of genomic data and the multiplicity of cell lines available, has enabled researchers to identify critical conserved pathways regulating self-renewal and identify markers that tightly correlate with the ES cell state. Comparison across species has suggested additional pathways likely to be important in long-term self-renewal of ES cells including heterochronic genes, microRNAs, genes involved in telomeric regulation, and polycomb repressors. In this review, we have discussed information on molecules known to be important in ES cell self-renewal or blastocyst development and highlighted known differences between mouse and human ES cells. We suggest that several additional pathways required for self-renewal remain to be discovered and these likely include genes involved in antisense regulation, microRNAs, as well as additional global repressive pathways and novel genes. We suggest that cross species comparisons using large-scale genomic analysis tools are likely to reveal conserved and divergent paths required for ES cell self-renewal and will allow us to derive ES lines from species and strains where this has been difficult.
Collapse
Affiliation(s)
- Mahendra Rao
- Stem Cell Section, Laboratory of Neurosciences, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
495
|
Cai J, Weiss ML, Rao MS. In search of "stemness". Exp Hematol 2004; 32:585-98. [PMID: 15246154 PMCID: PMC3279197 DOI: 10.1016/j.exphem.2004.03.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2004] [Revised: 03/22/2004] [Accepted: 03/25/2004] [Indexed: 01/02/2023]
Abstract
Stem cells have been identified and characterized in a variety of tissues. In this review we examine possible shared properties of stem cells. We suggest that irrespective of their lineal origin, stem cells have to respond in similar ways to regulate self-renewal and differentiation and it is likely that cell-cycle control, asymmetry/differentiation controls, cellular protective and DNA repair mechanisms, and associated apoptosis/senescence signaling pathways all might be expected to be more highly regulated in stem cells, likely by similar mechanisms. We review the literature to suggest a set of candidate stemness genes that may serve as universal stem cell markers. While we predict many similarities, we also predict that differences will exist between stem cell populations and that when transdifferentiation is considered genes expected to be both similar and different need to be examined.
Collapse
Affiliation(s)
- Jingli Cai
- Gerontology Research Center, Stem Cell Biology Unit/Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
496
|
Brandenberger R, Khrebtukova I, Thies RS, Miura T, Jingli C, Puri R, Vasicek T, Lebkowski J, Rao M. MPSS profiling of human embryonic stem cells. BMC DEVELOPMENTAL BIOLOGY 2004; 4:10. [PMID: 15304200 PMCID: PMC514702 DOI: 10.1186/1471-213x-4-10] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Accepted: 08/10/2004] [Indexed: 01/13/2023]
Abstract
Background Pooled human embryonic stem cells (hESC) cell lines were profiled to obtain a comprehensive list of genes common to undifferentiated human embryonic stem cells. Results Pooled hESC lines were profiled to obtain a comprehensive list of genes common to human ES cells. Massively parallel signature sequencing (MPSS) of approximately three million signature tags (signatures) identified close to eleven thousand unique transcripts, of which approximately 25% were uncharacterised or novel genes. Expression of previously identified ES cell markers was confirmed and multiple genes not known to be expressed by ES cells were identified by comparing with public SAGE databases, EST libraries and parallel analysis by microarray and RT-PCR. Chromosomal mapping of expressed genes failed to identify major hotspots and confirmed expression of genes that map to the X and Y chromosome. Comparison with published data sets confirmed the validity of the analysis and the depth and power of MPSS. Conclusions Overall, our analysis provides a molecular signature of genes expressed by undifferentiated ES cells that can be used to monitor the state of ES cells isolated by different laboratories using independent methods and maintained under differing culture conditions
Collapse
Affiliation(s)
- Ralph Brandenberger
- National Institute on Aging; GRC; Laboratory of Neuroscience, 5600 Nathan Shock Drive; Room 4E02; Baltimore, MD 21224, USA
| | - Irina Khrebtukova
- Lynx Therapeutics, Inc. 25861 Industrial Blvd., Hayward, CA 94545, USA
| | - R Scott Thies
- Geron Corporation, 230 Constitution Drive, Menlo Park, CA 94025, USA
| | - Takumi Miura
- National Institute on Aging; GRC; Laboratory of Neuroscience, 5600 Nathan Shock Drive; Room 4E02; Baltimore, MD 21224, USA
| | | | - Raj Puri
- Laboratory of Molecular Tumor Biology, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892
| | - Tom Vasicek
- Lynx Therapeutics, Inc. 25861 Industrial Blvd., Hayward, CA 94545, USA
| | - Jane Lebkowski
- Geron Corporation, 230 Constitution Drive, Menlo Park, CA 94025, USA
| | - Mahendra Rao
- National Institute on Aging; GRC; Laboratory of Neuroscience, 5600 Nathan Shock Drive; Room 4E02; Baltimore, MD 21224, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
497
|
Abstract
Embryonic stem (ES) cells can in theory produce all cell types of a living organism while renewing themselves with a stable genetic background. These unique features make ES cells a favorable tool for biomedical researches as well as a potential source for therapeutic application. A first step for approaching to ES cells is the directed differentiation to cells of interest, such as the neural cell lineage. Here, we summarize the up and down sides of each category of neural differentiation protocols that have so far been used in mouse and human ES cells, and introduce an efficient and plausible method used in our laboratory for derivation of neuroectodermal cells from human ES cells. This synthesis has led to our suggestions on issues for future design of neural differentiation protocols.
Collapse
Affiliation(s)
- Zhong-Wei Du
- Department of Anatomy, School of Medicine, Waisman Center, Wicell Institute, University of Wisconsin, Madison, WI 53705, USA
| | | |
Collapse
|
498
|
Zeng X, Miura T, Luo Y, Bhattacharya B, Condie B, Chen J, Ginis I, Lyons I, Mejido J, Puri RK, Rao MS, Freed WJ. Properties of Pluripotent Human Embryonic Stem Cells BG01 and BG02. Stem Cells 2004; 22:292-312. [PMID: 15153607 DOI: 10.1634/stemcells.22-3-292] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human ES (hES) cell lines have only recently been generated, and differences between human and mouse ES cells have been identified. In this manuscript we describe the properties of two human ES cell lines, BG01 and BG02. By immunocytochemistry and reverse transcription polymerase chain reaction, undifferentiated cells expressed markers that are characteristic of ES cells, including SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, and OCT-3/4. Both cell lines were readily maintained in an undifferentiated state and could differentiate into cells of all three germ layers, as determined by expression of beta-tubulin III neuron-specific molecule (ectoderm), cardiac troponin I (cardiomyocytes, mesoderm), and alpha-fetoprotein (endoderm). A large-scale microarray (16,659 genes) analysis identified 373 genes that were expressed at three-fold or higher levels in undifferentiated BG01 and BG02 cells as compared with pooled human RNA. Ninety-two of these genes were also highly expressed in four other hES lines (TE05, GE01, GE09, and pooled samples derived from GE01, GE09, and GE07). Included in the list are genes involved in cell signaling and development, metabolism, transcription regulation, and many hypothetical proteins. Two focused arrays designed to examine transcripts associated with stem cells and with the transforming growth factor-beta superfamily were employed to examine differentially expressed genes. Several growth factors, receptors, and components of signaling pathways that regulate embryonic development, in particular the nodal signaling pathway, were detected in both BG01 and BG02. These data provide a detailed characterization and an initial gene expression profile for the BG01 and BG02 human ES cell lines.
Collapse
Affiliation(s)
- Xianmin Zeng
- Development and Plasticity Section, Cellular Neurobiology Research Branch, National Institute on Drug Abuse, Department of Health and Human Services, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|