1
|
Ozawa M, Taguchi J, Katsuma K, Ishikawa-Yamauchi Y, Kikuchi M, Sakamoto R, Yamada Y, Ikawa M. Efficient simultaneous double DNA knock-in in murine embryonic stem cells by CRISPR/Cas9 ribonucleoprotein-mediated circular plasmid targeting for generating gene-manipulated mice. Sci Rep 2022; 12:21558. [PMID: 36513736 PMCID: PMC9748034 DOI: 10.1038/s41598-022-26107-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Gene targeting of embryonic stem (ES) cells followed by chimera production has been conventionally used for developing gene-manipulated mice. Although direct knock-in (KI) using murine zygote via CRISPR/Cas9-mediated genome editing has been reported, ES cell targeting still has merits, e.g., high throughput work can be performed in vitro. In this study, we first compared the KI efficiency of mouse ES cells with CRISPR/Cas9 expression vector and ribonucleoprotein (RNP), and confirmed that KI efficiency was significantly increased by using RNP. Using CRISPR/Cas9 RNP and circular plasmid with homologous arms as a targeting vector, knock-in within ES cell clones could be obtained efficiently without drug selection, thus potentially shortening the vector construction or cell culture period. Moreover, by incorporating a drug-resistant cassette into the targeting vectors, double DNA KI can be simultaneously achieved at high efficiency by a single electroporation. This technique will help to facilitate the production of genetically modified mouse models that are fundamental for exploring topics related to human and mammalian biology.
Collapse
Affiliation(s)
- Manabu Ozawa
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Jumpei Taguchi
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Kento Katsuma
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Yu Ishikawa-Yamauchi
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Mio Kikuchi
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Reiko Sakamoto
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Yasuhiro Yamada
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Masahito Ikawa
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.136593.b0000 0004 0373 3971Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871 Japan
| |
Collapse
|
2
|
Partial inhibition of differentiation associated with elevated protein levels of pluripotency factors in mouse embryonic stem cells expressing exogenous EGAM1N homeoprotein. J Biosci Bioeng 2015; 120:562-9. [DOI: 10.1016/j.jbiosc.2015.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 02/27/2015] [Accepted: 03/02/2015] [Indexed: 01/14/2023]
|
3
|
Wu S, Aksoy M, Shi J, Houbaviy HB. Evolution of the miR-290-295/miR-371-373 cluster family seed repertoire. PLoS One 2014; 9:e108519. [PMID: 25268927 PMCID: PMC4182485 DOI: 10.1371/journal.pone.0108519] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/21/2014] [Indexed: 12/20/2022] Open
Abstract
Expression of the mouse miR-290–295 cluster and its miR-371–373 homolog in human is restricted to early embryos, primordial germ cells, the germ line stem cell compartment of the adult testis and to stem cell lines derived from the early embryonic lineages. Sequencing data suggest considerable seed diversification between the seven homologous pre-miRNAs of miR-290–295 but it is not clear if all of the implied miR-290–295 seeds are also conserved in the human miR-371–373 cluster, which consists of only three homologous pre-miRNAs. By employing miRNA target reporters we show that most, if not all, seeds in miR-290–295 are represented in miR-371–373. In the mouse, pre-miR-290, pre-miR-292 and pre-miR-293 express subsets of the miRNA isoforms processed from the single human pre-miR-371. Comparison of the possible miR-290–295/miR-371–373 seed repertoires in placental mammals suggests a model for the evolution of this miRNA cluster family, which would be otherwise difficult to deduce based solely on pre-miRNA sequence comparisons. The conservation of co-expressed seeds that is characteristic of miR-290–295/miR-371–373 should be taken into account in models of the corresponding miRNA-target interaction networks.
Collapse
Affiliation(s)
- Shuang Wu
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Two Medical Center Drive, Stratford, NJ, United States of America
| | - Munevver Aksoy
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Two Medical Center Drive, Stratford, NJ, United States of America
| | - Jianting Shi
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Two Medical Center Drive, Stratford, NJ, United States of America
| | - Hristo Botev Houbaviy
- Department of Cell Biology, Rowan University School of Osteopathic Medicine, Two Medical Center Drive, Stratford, NJ, United States of America
- * E-mail:
| |
Collapse
|
4
|
Mezzanotte L, Aswendt M, Tennstaedt A, Hoeben R, Hoehn M, Löwik C. Evaluating reporter genes of different luciferases for optimized in vivo bioluminescence imaging of transplanted neural stem cells in the brain. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 8:505-13. [PMID: 24375906 DOI: 10.1002/cmmi.1549] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 05/07/2013] [Accepted: 05/10/2013] [Indexed: 11/05/2022]
Abstract
Bioluminescence imaging (BLI) has become the method of choice for optical tracking of cells in small laboratory animals. However, the use of luciferases from different species, depending on different substrates and emitting at distinct wavelengths, has not been optimized for sensitive neuroimaging. In order to identify the most suitable luciferase, this quantitative study compared the luciferases Luc2, CBG99, PpyRE9 and hRluc. Human embryonic kidney (HEK-293) cells and mouse neural stem cells were transduced by lentiviral vector-mediated transfer to express one of the four luciferases, together with copGFP. A T2A peptide linker promoted stoichiometric expression between both imaging reporters and the comparison of cell populations upon flow cytometry. Cell dilution series were used to determine highest BLI sensitivity in vitro for Luc2. However, Coelenterazine h-dependent hRluc signals clearly exceeded d-luciferin-dependent BLI in vitro. For the quantitative in vivo analysis, cells were transplanted into mouse brain and BLI was performed including the recording of emission kinetics and spectral characteristics. Differences in light kinetics were observed for d-luciferin vs Coelenterazine h. The emission spectra of Luc2 and PpyRE9 remained almost unchanged, while the emission spectrum of CBG99 became biphasic. Most importantly, photon emission decreased in the order of Luc2, CBG99, PpyRE9 to hRluc. The feasibility of combining different luciferases for dual color and dual substrate neuroimaging was tested and discussed. This investigation provides the first complete quantitative comparison of different luciferases expressed by neural stem cells. It results in a clear recommendation of Luc2 as the best luciferase selection for in vivo neuroimaging.
Collapse
Affiliation(s)
- Laura Mezzanotte
- Experimental Molecular Imaging, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
5
|
Hu K. All roads lead to induced pluripotent stem cells: the technologies of iPSC generation. Stem Cells Dev 2014; 23:1285-300. [PMID: 24524728 PMCID: PMC4046204 DOI: 10.1089/scd.2013.0620] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/12/2014] [Indexed: 12/26/2022] Open
Abstract
Generation of induced pluripotent stem cells (iPSCs) via the ectopic expression of reprogramming factors is a simple, advanced, yet often perplexing technology due to low efficiency, slow kinetics, and the use of numerous distinct systems for factor delivery. Scientists have used almost all available approaches for the delivery of reprogramming factors. Even the well-established retroviral vectors confuse some scientists due to different tropisms in use. The canonical virus-based reprogramming poses many problems, including insertional mutagenesis, residual expression and re-activation of reprogramming factors, uncontrolled silencing of transgenes, apoptosis, cell senescence, and strong immunogenicity. To eliminate or alleviate these problems, scientists have tried various other approaches for factor delivery and transgene removal. These include transient transfection, nonintegrating viral vectors, Cre-loxP excision of transgenes, excisable transposon, protein transduction, RNA transfection, microRNA transfection, RNA virion, RNA replicon, nonintegrating replicating episomal plasmids, minicircles, polycistron, and preintegration of inducible reprogramming factors. These alternative approaches have their own limitations. Even iPSCs generated with RNA approaches should be screened for possible transgene insertions mediated by active endogenous retroviruses in the human genome. Even experienced researchers may encounter difficulty in selecting and using these different technologies. This survey presents overviews of iPSC technologies with the intention to provide a quick yet comprehensive reference for both new and experienced reprogrammers.
Collapse
Affiliation(s)
- Kejin Hu
- Department of Biochemistry and Molecular Genetics, UAB Stem Cell Insitute, School of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
6
|
Yamato E, Bamba Y, Kamiya Y, Yagi K, Miyazaki JI. Analysis of the transcription factor cascade that induces endocrine and exocrine cell lineages from pancreatic progenitor cells using a polyoma-based episomal vector system. J Diabetes Investig 2014; 3:41-51. [PMID: 24843545 PMCID: PMC4014932 DOI: 10.1111/j.2040-1124.2011.00136.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Aims/Introduction: We recently established a strategy for isolating multipotential duct‐like cells, called pdx‐1‐positive pancreatic cell‐derived (PPPD) cells, from the pancreas. To analyze the molecular mechanisms of pancreatic cell differentiation, we introduced a polyoma‐based episomal vector system into PPPD cells. Materials and Methods: PPPD cells were stably transfected with a polyoma large T (PLT)‐expressing plasmid vector, which included the polyoma origin of replication, to generate PLT‐PPPD cells. Various cDNA for pancreas‐related transcription factors were subcloned into the expression plasmid pPyCAG, which included the polyoma origin of replication. PLT‐PPPD cells were stably transfected with the resulting plasmid vectors and then subjected to gene and protein expression analyses. Results: The coexpression of Mafa, Neurod1 and Ipf1 induced Ins1 and Ins2 expression in PLT‐PPPD cells. The forced expression of Pax6 alone induced the expression of glucagon. The coexpression of Neurod1 and Isl1 induced Ins2 and Sst expression. In contrast, the expression of Ptf1a and Foxa2 induced the expression of exocrine markers Cpa1 and Amy2. Transfections with multiple transcription factors showed that Isl1 is required for the differentiation of both insulin‐positive cells and somatostatin‐positive cells. In addition, Foxa2 induced the differentiation of glucagon‐positive cells and inhibited the differentiation of insulin‐positive and somatostatin‐positive cells. PLT‐PPPD cells allow episomal vector‐based gene expression and should be useful for studying the transcription factor cascades involved in the differentiation of pancreatic cell types in vitro. Conclusions: Our coexpression study showed novel critical roles for Isl1 and Foxa2 in the differentiation of PPPD cells into endocrine cells. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2011.00136.x, 2012)
Collapse
Affiliation(s)
- Eiji Yamato
- Stem Cell Regulation Research, Osaka University Graduate School of Medicine
| | - Yohei Bamba
- Stem Cell Regulation Research, Osaka University Graduate School of Medicine
| | - Yukimasa Kamiya
- Stem Cell Regulation Research, Osaka University Graduate School of Medicine ; Bio-functional Molecular Chemistry, Osaka University Graduate School of Pharmaceutical Sciences, Suita, Japan
| | - Kiyohito Yagi
- Bio-functional Molecular Chemistry, Osaka University Graduate School of Pharmaceutical Sciences, Suita, Japan
| | - Jun-Ichi Miyazaki
- Stem Cell Regulation Research, Osaka University Graduate School of Medicine
| |
Collapse
|
7
|
Shimamoto R, Amano N, Ichisaka T, Watanabe A, Yamanaka S, Okita K. Generation and characterization of induced pluripotent stem cells from Aid-deficient mice. PLoS One 2014; 9:e94735. [PMID: 24718089 PMCID: PMC3981863 DOI: 10.1371/journal.pone.0094735] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 03/18/2014] [Indexed: 12/15/2022] Open
Abstract
It has been shown that DNA demethylation plays a pivotal role in the generation of induced pluripotent stem (iPS) cells. However, the underlying mechanism of this action is still unclear. Previous reports indicated that activation-induced cytidine deaminase (Aid, also known as Aicda) is involved in DNA demethylation in several developmental processes, as well as cell fusion-mediated reprogramming. Based on these reports, we hypothesized that Aid may be involved in the DNA demethylation that occurs during the generation of iPS cells. In this study, we examined the function of Aid in iPS cell generation using Aid knockout (Aid−/−) mice expressing a GFP reporter under the control of a pluripotent stem cell marker, Nanog. By introducing Oct3/4, Sox2, Klf4 and c-Myc, Nanog-GFP-positive iPS cells could be generated from the fibroblasts and primary B cells of Aid−/− mice. Their induction efficiency was similar to that of wild-type (Aid+/+) iPS cells. The Aid−/− iPS cells showed normal proliferation and gave rise to chimeras, indicating their capacity for self-renewal and pluripotency. A comprehensive DNA methylation analysis showed only a few differences between Aid+/+ and Aid−/− iPS cells. These data suggest that Aid does not have crucial functions in DNA demethylation during iPS cell generation.
Collapse
Affiliation(s)
- Ren Shimamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Naoki Amano
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Tomoko Ichisaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, United States of America
| | - Keisuke Okita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
8
|
Nagashima F, Suzuki IK, Shitamukai A, Sakaguchi H, Iwashita M, Kobayashi T, Tone S, Toida K, Vanderhaeghen P, Kosodo Y. Novel and robust transplantation reveals the acquisition of polarized processes by cortical cells derived from mouse and human pluripotent stem cells. Stem Cells Dev 2014; 23:2129-42. [PMID: 24325299 DOI: 10.1089/scd.2013.0251] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Current stem cell technologies have enabled the induction of cortical progenitors and neurons from embryonic stem cells (ESCs) and induced pluripotent stem cells in vitro. To understand the mechanisms underlying the acquisition of apico-basal polarity and the formation of processes associated with the stemness of cortical cells generated in monolayer culture, here, we developed a novel in utero transplantation system based on the moderate dissociation of adherens junctions in neuroepithelial tissue. This method enables (1) the incorporation of remarkably higher numbers of grafted cells and (2) quantitative morphological analyses at single-cell resolution, including time-lapse recording analyses. We then grafted cortical progenitors induced from mouse ESCs into the developing brain. Importantly, we revealed that the mode of process extension depends on the extrinsic apico-basal polarity of the host epithelial tissue, as well as on the intrinsic differentiation state of the grafted cells. Further, we successfully transplanted cortical progenitors induced from human ESCs, showing that our strategy enables investigation of the neurogenesis of human neural progenitors within the developing mouse cortex. Specifically, human cortical cells exhibit multiple features of radial migration. The robust transplantation method established here could be utilized both to uncover the missing gap between neurogenesis from ESCs and the tissue environment and as an in vivo model of normal and pathological human corticogenesis.
Collapse
|
9
|
Intact structure of EGAM1 homeoproteins and basic amino acid residues in the common homeodomain of EGAM1 and EGAM1C contribute to their nuclear localization in mouse embryonic stem cells. J Biosci Bioeng 2013; 116:141-6. [PMID: 23510645 DOI: 10.1016/j.jbiosc.2013.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 02/13/2013] [Accepted: 02/13/2013] [Indexed: 11/22/2022]
Abstract
Recently, we identified the structurally related homeoproteins EGAM1, EGAM1N, and EGAM1C in both preimplantation mouse embryos and mouse embryonic stem (ES) cells. These EGAM1 homeoproteins act as positive or negative regulators of differentiation and cell growth in mouse ES cells, such that these proteins are considered transcriptional regulators. In this study, we investigated their nuclear localization and identified the amino acid residues crucial for the nuclear translocation of EGAM1 and EGAM1C. When expressed exogenously in pluripotent ES cells and somatic NIH3T3 cells, all EGAM1 homeoproteins localized to the nucleus. Analysis using the web-based tool PSORTII predicted a potential nuclear localization signal (NLS) motif, RKDLIRSWFITQRHR, in the homeodomain shared by EGAM1 and EGAM1C. The introduction of mutations, such as mutations from K or R, both basic amino acid residues, to A, in this potential NLS resulted in significant impairment of the nuclear localization of both EGAM1 and EGAM1C. In contrast, GFP fusion proteins of all the full-length EGAM1 homeoproteins failed to localize to the nucleus. These results, when taken together, suggest that basic amino acid residues in the common homeodomain of EGAM1 and EGAM1C and the intact structures of the EGAM1 homeoproteins contribute, at least in part, to the nuclear localization of these proteins in mouse ES cells.
Collapse
|
10
|
Serrano F, Calatayud CF, Blazquez M, Torres J, Castell JV, Bort R. Gata4 Blocks Somatic Cell Reprogramming By Directly Repressing Nanog. Stem Cells 2012; 31:71-82. [DOI: 10.1002/stem.1272] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 10/04/2012] [Indexed: 12/31/2022]
|
11
|
Guallar D, Pérez-Palacios R, Climent M, Martínez-Abadía I, Larraga A, Fernández-Juan M, Vallejo C, Muniesa P, Schoorlemmer J. Expression of endogenous retroviruses is negatively regulated by the pluripotency marker Rex1/Zfp42. Nucleic Acids Res 2012; 40:8993-9007. [PMID: 22844087 PMCID: PMC3467079 DOI: 10.1093/nar/gks686] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Rex1/Zfp42 is a Yy1-related zinc-finger protein whose expression is frequently used to identify pluripotent stem cells. We show that depletion of Rex1 levels notably affected self-renewal of mouse embryonic stem (ES) cells in clonal assays, in the absence of evident differences in expression of marker genes for pluripotency or differentiation. By contrast, marked differences in expression of several endogenous retroviral elements (ERVs) were evident upon Rex1 depletion. We demonstrate association of REX1 to specific elements in chromatin-immunoprecipitation assays, most strongly to muERV-L and to a lower extent to IAP and musD elements. Rex1 regulates muERV-L expression in vivo, as we show altered levels upon transient gain-and-loss of Rex1 function in pre-implantation embryos. We also find REX1 can associate with the lysine-demethylase LSD1/KDM1A, suggesting they act in concert. Similar to REX1 binding to retrotransposable elements (REs) in ES cells, we also detected binding of the REX1 related proteins YY1 and YY2 to REs, although the binding preferences of the two proteins were slightly different. Altogether, we show that Rex1 regulates ERV expression in mouse ES cells and during pre-implantation development and suggest that Rex1 and its relatives have evolved as regulators of endogenous retroviral transcription.
Collapse
Affiliation(s)
- D Guallar
- Regenerative Medicine Programme, IIS Aragón, Instituto Aragonés de Ciencias de Salud, Zaragoza, Avda. Gómez Laguna, 50009 Zaragoza, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Soma M, Iha M, Kihara Y, Sato S, Sato Y, Sato S, Mori Y, Sugawara S, Kasuga K, Kojima I, Kobayashi M. Preferential emergence of cell types expressing markers for primitive endoderm lineages in mouse embryonic stem cells expressing exogenous EGAM1 homeoprotein. J Biosci Bioeng 2012; 114:342-6. [PMID: 22608553 DOI: 10.1016/j.jbiosc.2012.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 04/16/2012] [Accepted: 04/18/2012] [Indexed: 11/17/2022]
Abstract
Embryonic stem (ES) cells have been considered as a valuable renewable source of materials in regenerative medicine. Recently, we identified the homeoprotein EGAM1 both in preimplantation mouse embryos and mouse ES cells. Expression of the Egam1 transcript and its encoded protein was detectable in differentiating mouse ES cells, while it was almost undetectable in undifferentiated cells. In the present study, in order to clarify the effect of forced expression of EGAM1 on the differentiation of mouse ES cells in vitro, transfectants expressing exogenous EGAM1 were generated. Egam1 transfectants promoted differentiation into cell types expressing Gata6, Gata4, Afp, or Plat, genes associated with emergence of the extra-embryonic endoderm lineages. On the other hand, Egam1 transfectants inhibited the expression of specific genes for the embryonic lineages, including Fgf5 (epiblast) and T (mesoderm), in addition to Cdx2, a specific gene for the extra-embryonic trophectoderm lineages. Changes in the percentage of cells recognizing by antibodies against specific marker proteins closely correlated with the expression patterns of their transcripts. Taken together, the results obtained in this study suggested that mouse ES cells expressing exogenous EGAM1 preferentially differentiate into extra-embryonic primitive endoderm lineages, rather than embryonic lineages or extra-embryonic trophectoderm lineages.
Collapse
Affiliation(s)
- Miki Soma
- Laboratory for Advanced Animal Cell Technology, Graduate School of Bioresource Sciences, Akita Prefectural University, Akita 010-0195, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Iha M, Watanabe M, Kihara Y, Sugawara S, Saito K, Soma M, Sato S, Mori Y, Kasuga K, Kojima I, Sasamura R, Murata J, Kobayashi M. Effect of ectopic expression of homeoprotein EGAM1C on the cell morphology, growth, and differentiation in a mouse embryonic stem cell line, MG1.19 cells. Reproduction 2012; 143:477-89. [DOI: 10.1530/rep-11-0379] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The homeoprotein EGAM1C was identified in preimplantation mouse embryos and embryonic stem (ES) cells. To explore the impact of EGAM1C on the hallmarks of mouse ES cells, MG1.19 cells stably expressing EGAM1C at levels similar to those in blastocysts were established using an episomal expression system. In the presence of leukemia inhibitory factor (+LIF), control transfectants with an empty vector formed flattened cell colonies, whileEgam1ctransfectants formed compacted colonies with increased E-CADHERIN expression. InEgam1ctransfectants, the cellular contents of POU5F1 (OCT4), SOX2, TBX3, and NANOG increased. Cell growth was accelerated in an undifferentiated state sustained by LIF and in the course of differentiation. During clonal proliferation, EGAM1C stabilized the undifferentiated state. In adherent culture conditions, EGAM1C partly inhibited the progression of differentiation at least within a 4-day culture period in the presence of retinoic acid by preventing the downregulation of LIF signaling with a robust increase in TBX3 expression. Conversely, EGAM1C enhanced the expression of lineage marker genesFgf5(epiblast),T(mesoderm),Gata6(primitive endoderm), andCdx2(trophectoderm) in −LIF conditions. In embryoid bodies expressing EGAM1C, the expression of marker genes for extraembryonic cell lineages, includingTpbpa(spongiotrophoblast) andPlat(parietal endoderm), increased. These results demonstrated that the ectopic expression of EGAM1C is capable of affecting the stabilization of an undifferentiated state and the progression of differentiation in MG1.19 ES cells, in addition to affecting cellular morphology and growth.
Collapse
|
14
|
Westerman BA, Braat AK, Taub N, Potman M, Vissers JHA, Blom M, Verhoeven E, Stoop H, Gillis A, Velds A, Nijkamp W, Beijersbergen R, Huber LA, Looijenga LHJ, van Lohuizen M. A genome-wide RNAi screen in mouse embryonic stem cells identifies Mp1 as a key mediator of differentiation. ACTA ACUST UNITED AC 2011; 208:2675-89. [PMID: 22143885 PMCID: PMC3244037 DOI: 10.1084/jem.20102037] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite intense investigation of intrinsic and extrinsic factors that regulate pluripotency, the process of initial fate commitment of embryonic stem (ES) cells is still poorly understood. We used a genome-wide short hairpin RNA screen in mouse ES cells to identify genes that are essential for initiation of differentiation. Knockdown of the scaffolding protein Mek binding protein 1 (Mp1, also known as Lamtor3 or Map2k1ip1) stimulated self-renewal of ES cells, blocked differentiation, and promoted proliferation. Fibroblast growth factor 4 (FGF4) signaling is required for initial fate commitment of ES cells. Knockdown of Mp1 inhibited FGF4-induced differentiation but did not alter FGF4-driven proliferation. This uncoupling of differentiation and proliferation was also observed when oncogenic Ras isoforms were overexpressed in ES cells. Knockdown of Mp1 redirected FGF4 signaling from differentiation toward pluripotency and up-regulated the pluripotency-related genes Esrrb, Rex1, Tcl1, and Sox2. We also found that human germ cell tumors (GCTs) express low amounts of Mp1 in the invasive embryonic carcinoma and seminoma histologies and higher amounts of Mp1 in the noninvasive carcinoma in situ precursor and differentiated components. Knockdown of Mp1 in invasive GCT cells resulted in resistance to differentiation, thereby showing a functional role for Mp1 both in normal differentiation of ES cells and in germ cell cancer.
Collapse
Affiliation(s)
- Bart A Westerman
- Division of Molecular Genetics, the Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Liu JF, Chen YM, Yang JJ, Kurokawa T, Kakugo A, Yamamoto K, Gong JP. Dynamic Behavior and Spontaneous Differentiation of Mouse Embryoid Bodies on Hydrogel Substrates of Different Surface Charge and Chemical Structures. Tissue Eng Part A 2011; 17:2343-57. [DOI: 10.1089/ten.tea.2011.0034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Jian Fang Liu
- Department of Biological Sciences, Graduate School of Science, Hokkaido University, Sapporo, Japan
| | - Yong Mei Chen
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Jing Jing Yang
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Takayuki Kurokawa
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
- Creative Research Institution Sosei, Hokkaido University, Sapporo, Japan
| | - Akira Kakugo
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Kimiko Yamamoto
- Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Jian Ping Gong
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
16
|
Abujarour R, Efe J, Ding S. Genome-wide gain-of-function screen identifies novel regulators of pluripotency. Stem Cells 2010; 28:1487-97. [PMID: 20629179 DOI: 10.1002/stem.472] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Pluripotent stem cells are characterized by the capacity to self-renew and to differentiate into all the cell types of the body. To identify novel regulators of pluripotency, we screened cDNA libraries (>30,000 clones) in P19 embryonal carcinoma cells for factors that modulate the expression of a luciferase reporter driven by the promoter of the pluripotency master regulator Nanog. Ninety confirmed hits activated the reporter and 14 confirmed hits inhibited the reporter by more than two-fold. The identified hits were evaluated by gain- and loss-of-functions approaches. The reporter-activating hits Timp2, Hig2, and Mki67ip promoted embryonic stem (ES) cell self-renewal when episomally overexpressed in ES cells, whereas the reporter-inhibiting hits PU.1/Spi1, Prkaca, and Jun induced differentiation of ES cells. Conversely, the knockdown of the activating hits Timp2, Mki67ip, Esrrg, and Dusp7 in ES cells induced differentiation, whereas the knockdown of the reporter-inhibiting hit PU.1/Spi1 led to inhibition of differentiation. One of the novel hits, the RNA-binding protein Mki67ip was further characterized, and found to be overexpressed in ES cells and in early development and downregulated during differentiation. The knockdown of Mki67ip led to the differentiation of ES cells, decreased growth rate, reduction in pluripotency markers, and induction of lineage-specific markers. In addition, colocalization and coimmunoprecipitation experiments suggest that Mki67ip promotes ES cell self-renewal via a mechanism involving nucleophosmin, a multifunctional nucleolar protein upregulated in stem cells and cancer.
Collapse
Affiliation(s)
- Ramzey Abujarour
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | |
Collapse
|
17
|
The transcription factor TCFAP2C/AP-2gamma cooperates with CDX2 to maintain trophectoderm formation. Mol Cell Biol 2010; 30:3310-20. [PMID: 20404091 DOI: 10.1128/mcb.01215-09] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In mammals, cell lineage specification is established at the blastocyst stage. At this stage, transcription factor Cdx2 represses pluripotency genes, thus promoting extraembryonic trophoblast fate. Recently, transcription factor Gata3 was shown to act in a parallel pathway in promoting trophoblast cell fate, suggesting that there are more factors working in the trophoblast lineage. Here, we report that the transcription factor Tcfap2c is expressed at a high level in the trophectoderm and is able to induce trophoblast fate in embryonic stem cells. Trophoblast fate induced by Tcfap2c does not require Cdx2 and vice versa, suggesting that the molecules act in alternative pathways. However, both Tcfap2c and Cdx2 are required for the upregulation of Elf5, a marker of trophoblast stem cell maintenance, suggesting that both factors are required for stable trophoblast induction. Tcfap2c-induced trophoblast-like cells are stable in long-term culture, indicating that they are capable of self-renewal. Tcfap2c-controlled trophoblast maintenance involves the induction of Cdx2 and the repression of the pluripotency factor Nanog. Tcfap2c-induced trophoblast-like cells differentiate to trophoblast derivatives in vitro and contribute to the trophectoderm in blastocysts in vivo. Taken together, these observations suggest that Tcfap2c and Cdx2 cooperate to override the pluripotency program and establish the extraembryonic trophoblast maintenance program in murine embryos.
Collapse
|
18
|
Abstract
Mouse embryonic stem (mES) cells are pluripotent stem cells derived from pre-implantation embryos. They are regarded as an essential tool for studying mouse development, as they provide a means for generating knock-out mouse lines. This, however, is not the sole utility of the mES cell system. They undergo differentiation in culture, mimicking the morphological differentiation of peri-implantation embryos from epiblast to egg-cylinder stage. Moreover, they retain the capacity to respond to triggers of differentiation toward trophectoderm and primitive endoderm by forced activation. For these reasons, mES cells can be regarded as a useful tool for analyzing molecular mechanisms underlying early mouse development.
Collapse
Affiliation(s)
- Hitoshi Niwa
- Laboratory for Pluripotent Cell Studies, RIKEN Center for Developmental Biology (CDB), Kobe, Hyogo, Japan.
| |
Collapse
|
19
|
Saito K, Abe H, Nakazawa M, Irokawa E, Watanabe M, Hosoi Y, Soma M, Kasuga K, Kojima I, Kobayashi M. Cloning of complementary DNAs encoding structurally related homeoproteins from preimplantation mouse embryos: their involvement in the differentiation of embryonic stem cells. Biol Reprod 2009; 82:687-97. [PMID: 20018908 DOI: 10.1095/biolreprod.108.075697] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
During the preimplantation development of mouse embryos between the 4-cell to 8-cell stage and the morula stage, when the first irreversible segregation of cell fates proceeds into the pluripotent inner cell mass (progenitor cells to form the fetus) and the trophectoderm (to form the placenta) of blastocysts, pluripotency-maintaining and differentiation-inducing genes are expressed to coordinately regulate cell fates. Three structurally related cDNAs (Crxos1, Crxos1 sv2, and Crxos1 tv3) that exhibited concomitant elevated expression during this critical period were identified by subtractive cDNA cloning. CRXOS1 contains two homeodomains, while CRXOS1 sv2 and CRXOS1 tv3 each contain one of the homeodomains included in CRXOS1. Crxos1, Crxos1 sv2, and Crxos1 tv3 were expressed differentially during in vitro embryonic stem (ES) cell differentiation. Even under differentiation-inducing conditions, forced expression of Crxos1 sv2 inhibited the differentiation of ES cells. In contrast, under conditions that promote self-renewal of ES cells, forced expression of Crxos1 induced differentiation. Forced expression of Crxos1 resulted in induction of Gata4 but in repression of T, probably indicating that Crxos1 promotes the differentiation of ES cells into primitive endoderm, while inhibiting differentiation into mesoderm. On the other hand, no apparent effects of forced expression of Crxos1 tv3 were observed. Taken together, it was concluded that these transcripts encoding homeoproteins are capable of regulating the maintenance and/or differentiation of mouse ES cells and likely regulate that of preimplantation embryos.
Collapse
Affiliation(s)
- Koichi Saito
- Department of Biotechnology, Faculty of Bioresource Sciences, Akita Prefectural University, Akita, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
CrxOS maintains the self-renewal capacity of murine embryonic stem cells. Biochem Biophys Res Commun 2009; 390:1129-35. [PMID: 19800316 DOI: 10.1016/j.bbrc.2009.09.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 09/23/2009] [Indexed: 01/14/2023]
Abstract
Embryonic stem (ES) cells maintain pluripotency by self-renewal. Several homeoproteins, including Oct3/4 and Nanog, are known to be key factors in maintaining the self-renewal capacity of ES cells. However, other genes required for the mechanisms underlying this process are still unclear. Here we report the identification by in silico analysis of a homeobox-containing gene, CrxOS, that is specifically expressed in murine ES cells and is essential for their self-renewal. ES cells mainly express the short isoform of endogenous CrxOS. Using a polyoma-based episomal expression system, we demonstrate that overexpression of the CrxOS short isoform is sufficient for maintaining the undifferentiated morphology of ES cells and stimulating their proliferation. Finally, using RNA interference, we show that CrxOS is essential for the self-renewal of ES cells, and provisionally identify foxD3 as a downstream target gene of CrxOS. To our knowledge, ours is the first delineation of the physiological role of CrxOS in ES cells.
Collapse
|
21
|
Kobayashi T, Mizuno H, Imayoshi I, Furusawa C, Shirahige K, Kageyama R. The cyclic gene Hes1 contributes to diverse differentiation responses of embryonic stem cells. Genes Dev 2009; 23:1870-5. [PMID: 19684110 DOI: 10.1101/gad.1823109] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stem cells do not all respond the same way, but the mechanisms underlying this heterogeneity are not well understood. Here, we found that expression of Hes1 and its downstream genes oscillate in mouse embryonic stem (ES) cells. Those expressing low and high levels of Hes1 tended to differentiate into neural and mesodermal cells, respectively. Furthermore, inactivation of Hes1 facilitated neural differentiation more uniformly at earlier time. Thus, Hes1-null ES cells display less heterogeneity in both the differentiation timing and fate choice, suggesting that the cyclic gene Hes1 contributes to heterogeneous responses of ES cells even under the same environmental conditions.
Collapse
Affiliation(s)
- Taeko Kobayashi
- Institute for Virus Research, Kyoto University, Kyoto, Japan.
| | | | | | | | | | | |
Collapse
|
22
|
Kanemura M, Abe M, Ueda M, Ueki M, Awaya A, Sato Y. MS-818 Accelerates Mobilization of Endothelial Progenitor Cells and Differentiation to Endothelial Cells. ACTA ACUST UNITED AC 2009; 11:221-30. [PMID: 15763941 DOI: 10.1080/10623320490904089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
MS-818 that is a synthetic pyrimidine compound and shown to have neurotrophic actions, enhanced basic fibroblast growth factor (bFGF)-induced angiogenesis in vivo. However, the mechanism and whether MS-818 affects endothelial cells (ECs) directly is not known. Here, the authors investigated whether MS-818 alone could induce angiogenesis and tried to clarify the mechanism of neovascularization by MS-818 in terms of angiogenesis and vasculogenesis. The authors show that MS-818 affects ECs directly and induces migration of and tube formation by ECs in vitro (angiogenesis). Furthermore, the authors demonstrate that MS-818 mobilizes endothelial progenitor cells (EPCs) from the bone marrow and potentiates their differentiation to ECs (vasculogenesis). The effect of MS-818 on the endothelial differentiation was further confirmed with an in vitro differentiation system using mouse embryonic stem cells. MS-818 activates the mitogen-activated protein kinase (MAPK) pathway but not the phosphoinositol 3-kinase (PI3K)-Akt pathway in ECs. These results indicate that MS-818, a synthetic compound, promotes both angiogenesis and vasculogenesis.
Collapse
Affiliation(s)
- Masanori Kanemura
- Department of Vascular Biology, Institute of Development, Aging, and Cancer, Tohoku University, Seiryo-machi, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
23
|
Dickkopf (Dkk) 1 promotes the differentiation of mouse embryonic stem cells toward neuroectoderm. In Vitro Cell Dev Biol Anim 2008; 45:185-93. [PMID: 19057969 DOI: 10.1007/s11626-008-9157-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Accepted: 10/16/2008] [Indexed: 12/31/2022]
Abstract
Wnt signaling has been demonstrated to have extensive roles during embryogenesis. The Wnt family is highly conserved. In mice, there are 19 Wnt genes. Dickkopf (Dkk), through its interactions with Wnt co-receptors, low-density lipoprotein receptor-related protein (LRP), Frizzled and Kremen, can act as a negative regulator to block the Wnt-signaling pathway. There are four Dkk genes in the human genome, and three in that of the mouse. Dkk1 is involved in a variety of craniofacial developmental processes and behaves as a strong head inducer and limb regulator. Dkk1 mutant mice are embryonic-lethal. Here, we investigated the effects of Dkk1 on the differentiation of murine ESCs in both the ESC and embryoid body (EB) states. The results demonstrate that Dkk1 overexpression can initiate the differentiation program of ESCs toward neuroectoderm. We believe this finding can augment our understanding of mouse ESC differentiation.
Collapse
|
24
|
Abstract
Devoted teachers and mentors during early childhood and adolescence nurtured my ambition to become a scientist, but it was not until I actually began doing experiments in college and graduate school that I was confident about that choice and of making it a reality. During my postdoctoral experiences and thereafter, I made several significant advances, most notably the discovery of the then novel acyl- and aminoacyl adenylates: the former as intermediates in fatty acyl coenzyme A (CoA) formation and the latter as precursors to aminoacyl tRNAs. In the early 1970s, my research changed from a focus on transcription and translation in Escherichia coli to the molecular genetics of mammalian cells. To that end, my laboratory developed a method for creating recombinant DNAs that led us and others, over the next two decades, to create increasingly sophisticated ways for introducing “foreign” DNAs into cultured mammalian cells and to target modifications of specific chromosomal loci. Circumstances surrounding that work drew me into the public policy debates regarding recombinant DNA practices. As an outgrowth of my commitment to teaching, I coauthored several textbooks on molecular genetics and a biography of George Beadle. The colleagues, students, and wealth of associates with whom I interacted have made being a scientist far richer than I can have imagined.
Collapse
Affiliation(s)
- Paul Berg
- Cahill Professor in Cancer Research and Biochemistry, Emeritus, Stanford University Medical Center, Stanford, California 94305-5020
| |
Collapse
|
25
|
Zhang C, Ye X, Zhang H, Ding M, Deng H. GATA factors induce mouse embryonic stem cell differentiation toward extraembryonic endoderm. Stem Cells Dev 2007; 16:605-13. [PMID: 17784834 DOI: 10.1089/scd.2006.0077] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The GATA family of transcription factors are implicated in early embryonic development. There are six factors in this family in vertebrates. GATA4 and GATA6 have been demonstrated to induce mouse embryonic stem (mES) cells differentiation toward extraembryonic endoderm (ExE). We investigated the effect of GATA3 on the differentiation of mES cells both in the ES cell and in the embryoid body (EB) states. The results demonstrate that GATA3 overexpression can initiate the ES cell differentiation program toward ExE. Furthermore, overexpression of GATA1 and GATA2 in ES cells and EBs resulted in similar effects. We believe this finding can augment our understanding of mouse ES cell differentiation.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Cell Biology and Genetics, College of Life Sciences, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
26
|
Samuel MS, Lundgren-May T, Ernst M. Identification of putative targets of DNA (cytosine-5) methylation-mediated transcriptional silencing using a novel conditionally active form of DNA methyltransferase 3a. Growth Factors 2007; 25:426-36. [PMID: 18365873 DOI: 10.1080/08977190801931081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Aberrant DNA methylation of gene promoters is a recurrent finding associated with diseases such as cancer and inflammation, and is thought to contribute to disease through its role in transcriptional repression. Indeed, recent evidence suggests that DNA (cytosine-5) methyltransferases (DNMTs) may mediate the activity of factors promoting cell growth. Here, we utilise a novel experimental system for the conditional and reversible activation of a de novo DNMT by constructing a steroid-hormone analogue activated version, Dnmt3a-mERtrade mark. Following treatment with the oestrogen analogue 4-hydroxy tamoxifen of murine embryonic stem cells expressing this protein, we have identified by microarray analysis, several potential targets of Dnmt3a mediated transcriptional repression including the cancer associated genes Ssx2ip, Hmga1 and Wrnip. These results were validated using quantitative reverse transcriptase PCR and we confirm the biological significance of these in vitro observations by demonstrating a reduction in mRNA transcripts of the same genes within the intestinal epithelium of cancer-prone transgenic knock-in mutant mice over-expressing Dnmt3a throughout the intestinal epithelium.
Collapse
Affiliation(s)
- Michael S Samuel
- Ludwig Institute for Cancer Research, P. O. Royal Melbourne Hospital, Parkville, Vic., Australia
| | | | | |
Collapse
|
27
|
Shimoda M, Kanai-Azuma M, Hara K, Miyazaki S, Kanai Y, Monden M, Miyazaki JI. Sox17 plays a substantial role in late-stage differentiation of the extraembryonic endoderm in vitro. J Cell Sci 2007; 120:3859-69. [DOI: 10.1242/jcs.007856] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sox17 is a Sry-related HMG-box transcription factor developmentally expressed in both the definitive endoderm and extraembryonic endoderm (ExE). Although Sox17–/– mouse embryos have a defective definitive gut endoderm, their developing ExE is morphologically intact. Here, we aimed to investigate the role of Sox17 in ExE development by using an in vitro differentiation system of embryonic stem cells (ESCs). Although forced Sox17 expression in ESCs did not affect ExE commitment, it facilitated the differentiation of ESC-derived primitive endoderm cells into visceral and parietal endoderm cells. This event was inhibited by the forced expression of Nanog, a negative regulator of differentiation of ESCs into the ExE. Although Sox17–/– ESCs could differentiate into primitive endoderm cells, further differentiation was severely impaired. These results indicate a substantial involvement of Sox17 in the late stage of ExE differentiation in vitro. Furthermore, the expression of Sox7 – another Sox factor, concomitantly expressed with Sox17 in the developing ExE – was suppressed during the in vitro differentiation of Sox17–/– ESCs, but it was maintained at a high level in the extraembryonic tissues of Sox17–/– embryos. These findings possibly explain the discrepancy between the ExE phenotype derived from Sox17–/– ESCs and that of Sox17–/– embryos.
Collapse
Affiliation(s)
- Masafumi Shimoda
- Department of Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masami Kanai-Azuma
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Kenshiro Hara
- Department of Veterinary Anatomy, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo 113-8657, Japan
| | - Satsuki Miyazaki
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo 113-8657, Japan
| | - Morito Monden
- Department of Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jun-ichi Miyazaki
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
28
|
Sugiura M, Nagaoka M, Yabuuchi H, Akaike T. Overexpression of MCT8 enhances the differentiation of ES cells into neural progenitors. Biochem Biophys Res Commun 2007; 360:741-5. [PMID: 17624308 DOI: 10.1016/j.bbrc.2007.06.109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Accepted: 06/21/2007] [Indexed: 11/23/2022]
Abstract
Embryonic stem (ES) cell differentiation is regulated by cytokines and growth factors, as well as small-compound chemicals incorporated into cells by transporter proteins. Little is known regarding the effect of transporters on ES cell differentiation. This study focused on the effect of transporters during the neural-lineage differentiation of ES cells. Among the 27 types of SLC family transporters, MCT8 expression was coincident with that of neural stem cell markers, and the overexpression of MCT8 accelerated the differentiation into neural cells. These results suggested that the transporters and their substrates also play a crucial role in the regulation of ES cell differentiation.
Collapse
Affiliation(s)
- Mika Sugiura
- Department of Biomolecular Engineering, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259-B-57 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | |
Collapse
|
29
|
Adachi K, Soeta-Saneyoshi C, Sagara H, Iwakura Y. Crucial role of Bysl in mammalian preimplantation development as an integral factor for 40S ribosome biogenesis. Mol Cell Biol 2007; 27:2202-14. [PMID: 17242206 PMCID: PMC1820511 DOI: 10.1128/mcb.01908-06] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 11/15/2006] [Accepted: 01/04/2007] [Indexed: 02/03/2023] Open
Abstract
Blastocyst formation during mammalian preimplantation development is a unique developmental process that involves lineage segregation between the inner cell mass and the trophectoderm. To elucidate the molecular mechanisms underlying blastocyst formation, we have functionally screened a subset of preimplantation embryo-associated transcripts by using small interfering RNA (siRNA) and identified Bysl (bystin-like) as an essential gene for this process. The development of embryos injected with Bysl siRNA was arrested just prior to blastocyst formation, resulting in a defect in trophectoderm differentiation. Silencing of Bysl by using an episomal short hairpin RNA expression vector inhibited proliferation of embryonic stem cells. Exogenously expressed Bysl tagged with a fluorescent protein was concentrated in the nucleolus with a diffuse nucleoplasmic distribution. Furthermore, the loss of Bysl function by using RNA interference or dominant negative mutants caused defects in 40S ribosomal subunit biogenesis. These findings provide evidence for a crucial role of Bysl as an integral factor for ribosome biogenesis and suggest a critical dependence of blastocyst formation on active translation machinery.
Collapse
Affiliation(s)
- Kenjiro Adachi
- Institute of Medical Science, University of Tokyo, Minato-ku, Toyko 108-8639, Japan
| | | | | | | |
Collapse
|
30
|
Intravital imaging of fluorescent markers and FRET probes by DNA tattooing. BMC Biotechnol 2007; 7:2. [PMID: 17201912 PMCID: PMC1779781 DOI: 10.1186/1472-6750-7-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 01/03/2007] [Indexed: 11/13/2022] Open
Abstract
Background Advances in fluorescence microscopy and mouse transgenesis have made it possible to image molecular events in living animals. However, the generation of transgenic mice is a lengthy process and intravital imaging requires specialized knowledge and equipment. Here, we report a rapid and undemanding intravital imaging method using generally available equipment. Results By DNA tattooing we transfect keratinocytes of living mice with DNA encoding fluorescent biosensors. Subsequently, the behavior of individual cells expressing these biosensors can be visualized within hours and using conventional microscopy equipment. Using this "instant transgenic" model in combination with a corrected coordinate system, we followed the in vivo behavior of individual cells expressing either FRET- or location-based biosensors for several days. The utility of this approach was demonstrated by assessment of in vivo caspase-3 activation upon induction of apoptosis. Conclusion This "instant skin transgenic" model can be used to follow the in vivo behavior of individual cells expressing either FRET- or location-based probes for several days after tattooing and provides a rapid and inexpensive method for intravital imaging in murine skin.
Collapse
|
31
|
Abe M, Sato Y. Puromycin insensitive leucyl-specific aminopeptidase (PILSAP) is required for the development of vascular as well as hematopoietic system in embryoid bodies. Genes Cells 2006; 11:719-29. [PMID: 16824192 DOI: 10.1111/j.1365-2443.2006.00978.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We have shown that puromycin insensitive leucyl-specific aminopeptidase (PILSAP) is required for regulation of angiogenesis. However, it remains unclear whether PILSAP plays a role in endothelial cell (EC) differentiation. We examined the role of PILSAP by using an embryoid bodies (EBs) culture system. Fms-like tyrosine kinase-1 (Flk-1) showed two expression peaks on days 4 and 10 of culture. These two peaks represent populations of mesodermal precursors and mature ECs, respectively. Endothelial markers such as VE-cadherin, CD34, CD31 and Tie2 followed the first peak of Flk-1. Interestingly, the expression of PILSAP showed a pattern similar to that of Flk-1. ES cells transfected with mutant PILSAP (mtPILSAP) cDNA of a dominant negative activity organized less vascular structure and showed decreased levels of vascular lineage markers. The similar results were obtained in EBs treated with leucinethiol, a specific inhibitor of leucine aminopeptidase or siRNA for PILSAP. However, Flk-1 expression was unaffected on day 4. The expression of markers for hematopoietic lineage and muscle cells in mtPILSAP-EBs was also reduced. These results suggest that although PILSAP may not function in the initial generation of Flk-1 positive mesodermal precursors, it dose play a role in growth of vascular, hematopoietic, and muscular lineage population from those precursors.
Collapse
Affiliation(s)
- Mayumi Abe
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | | |
Collapse
|
32
|
Pritsker M, Ford NR, Jenq HT, Lemischka IR. Genomewide gain-of-function genetic screen identifies functionally active genes in mouse embryonic stem cells. Proc Natl Acad Sci U S A 2006; 103:6946-51. [PMID: 16621925 PMCID: PMC1458999 DOI: 10.1073/pnas.0509861103] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Indexed: 11/18/2022] Open
Abstract
Embryonic stem (ES) cells hold great promise for the future of medicine. To elucidate the molecular mechanisms that control ES cell self-renewal and differentiation, a comprehensive knowledge of the molecules involved in these processes is required. Here we describe an effective approach for genomewide identification of functionally active genes in ES cells. This approach combines genetic screens based on cDNA libraries with microarray detection methods to permit high-throughput functional analyses. We implement this strategy to identify genes whose overexpression can maintain phenotypic properties of undifferentiated mouse ES cells under differentiation-inducing conditions, specifically in the absence of leukemia inhibitory factor. The identified genes encode a variety of regulatory proteins whose function in ES cells was previously unknown. Moreover, our approach is capable of detecting genes whose overexpression promote differentiation or cell death. Overall, our studies establish a methodology for highly sensitive identification of genes that confer particular phenotypes on ES cells.
Collapse
Affiliation(s)
- Moshe Pritsker
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Nicole R. Ford
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Harry T. Jenq
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Ihor R. Lemischka
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
33
|
Friel R, Fisher D, Hook L. Embryonic stem cell technology: applications and uses in functional genomic studies. STEM CELL REVIEWS 2006; 2:31-5. [PMID: 17142884 DOI: 10.1007/s12015-006-0006-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 05/12/2023]
Abstract
In this postgenomic era, the role of functional genomics is becoming increasingly important and playing a key role in this field are embryonic stem cells. These cells are capable of proliferating indefinitely in a pluripotent state and have the potential to differentiate into all somatic cell types. Through a combination of their ease of genetic manipulation and directed in vitro differentiation they have proved themselves to be an extremely valuable tool in functional genomics. Here, some of their applications in functional genomic studies are discussed.
Collapse
Affiliation(s)
- Ruairi Friel
- Stem Cell Sciences Limited, Roger Land Building,West Mains Road, Edinburgh EH9 3JQ, UK.
| | | | | |
Collapse
|
34
|
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: 463] [Impact Index Per Article: 23.2] [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
|
35
|
Suzuki H, Watabe T, Kato M, Miyazawa K, Miyazono K. Roles of vascular endothelial growth factor receptor 3 signaling in differentiation of mouse embryonic stem cell–derived vascular progenitor cells into endothelial cells. Blood 2005; 105:2372-9. [PMID: 15561887 DOI: 10.1182/blood-2004-07-2547] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AbstractVascular endothelial growth factor receptor 2 (VEGFR2/Flk-1)–positive cells derived from embryonic stem (ES) cells serve as vascular progenitors, which differentiate into endothelial cells (ECs) in the presence of VEGF-A. VEGFR3/Flt-4 (fms-like tyrosine kinase 4) signaling is known to be important for the development of lymphatic endothelial cells (LECs). To elucidate the roles of VEGFR3 signaling in the differentiation of vascular progenitor cells into ECs, we introduced various types of VEGFR3 cDNAs into mouse ES cells. VEGF-C, a ligand for VEGFR2 and VEGFR3, stimulated the endothelial differentiation of the VEGFR2+ cells transfected with the VEGFR3 cDNA but not those transfected with kinasenegative mutants of VEGFR3. The VEGFR3-transfected ECs exhibited high expression levels of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), one of the markers of LECs, and showed efficient binding of hyaluronan. VEGF-C(C152S), which is able to activate VEGFR3 but not VEGFR2, failed to induce the endothelial differentiation of mock- and VEGFR3-transfected VEGFR2+ cells, suggesting the essential role of VEGFR2 signaling for endothelial differentiation. Furthermore, kinase-negative mutants of VEGFR3 prevented the VEGF-C–mediated endothelial differentiation of the vascular progenitor cells. Thus, VEGFR2 signaling is required for the endothelial differentiation of mouse ES cells induced by VEGF-C, and VEGFR3 signaling may confer lymphatic endothelial-like phenotypes to ECs.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | |
Collapse
|
36
|
Masui S, Shimosato D, Toyooka Y, Yagi R, Takahashi K, Niwa H. An efficient system to establish multiple embryonic stem cell lines carrying an inducible expression unit. Nucleic Acids Res 2005; 33:e43. [PMID: 15741176 PMCID: PMC552969 DOI: 10.1093/nar/gni043] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2004] [Revised: 02/10/2005] [Accepted: 02/10/2005] [Indexed: 11/23/2022] Open
Abstract
The growing use of mouse embryonic stem (ES) cells in research emphasizes their importance in studies of molecular mechanisms that maintain pluripotency and direct cellular differentiation. Although systems for regulatable transgene expression are essential for fine analysis of cellular processes at the molecular level, a strategy for the establishment of multiple ES cell lines carrying any of these systems has not yet been described. Here, we report our development of the ROSA-TET system, an effective system for the establishment of multiple ES cell lines carrying a tetracycline (Tc)-regulatable transgene at the Gt (ROSA)26asSor (ROSA26) locus. This system contains a knock-in step of a construct carrying both loxP and its mutant sequences into the ROSA26 locus, followed by a subsequent exchange step that introduces a cDNA to be Tc-regulated to the locus using the recombinase-mediated cassette exchange reaction. Both steps are demonstrated to give desired clones with high efficiency, suggesting that this system can be introduced readily into any ES cell lines, leading to the simultaneous establishment of multiple cell lines carrying different Tc-regulated cDNAs. We believe that use of this system will strongly accelerate molecular biological research using ES cells.
Collapse
Affiliation(s)
- Shinji Masui
- Laboratory for Pluripotent Cell Studies, RIKEN Center for Developmental Biology Minatojima-minamimachi 2-2-3, Kobe, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Zhao S, Nichols J, Smith AG, Li M. SoxB transcription factors specify neuroectodermal lineage choice in ES cells. Mol Cell Neurosci 2005; 27:332-42. [PMID: 15519247 DOI: 10.1016/j.mcn.2004.08.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2004] [Revised: 07/22/2004] [Accepted: 08/03/2004] [Indexed: 01/03/2023] Open
Abstract
Knowledge of lineage decision machinery in pluripotent embryonic stem (ES) cells may shed light on the process of germ layer segregation in the mammalian embryo and enable directed differentiation in vitro for biomedical applications. We have investigated the contribution of Class B1 Sox transcription factors to lineage choice during ES cell differentiation. We report that forced expression of Sox1 or Sox2 did not impair propagation of undifferentiated ES cells, but upon release from self-renewal promoted differentiation into neuroectoderm at the expense of mesoderm and endoderm. The efficient specification of a primary lineage by transcription factor manipulation provides a paradigm for instructing differentiation of ES cells for biopharmaceutical screening and cell therapy applications.
Collapse
Affiliation(s)
- Suling Zhao
- Institute for Stem Cell Research, University of Edinburgh, Edinburgh, EH9 3JQ, United Kingdom
| | | | | | | |
Collapse
|
38
|
Abstract
Embryonic stem (ES) cells are typically derived from the inner cell mass of the preimplantation blastocyst and can both self-renew and differentiate into all the cells and tissues of the embryo. Because they are pluripotent, ES cells have been used extensively to analyze gene function in development via gene targeting. The embryonic stem cell is also an unsurpassed starting material to begin to understand a critical, largely inaccessible period of development. If their differentiation could be controlled, they would also be an important source of cells for transplantation to replace cells lost through disease or injury or to replace missing hormones or genes. Traditionally, ES cells have been differentiated in suspension culture as embryoid bodies, named because of their similarity to the early postimplantation-staged embryo. Unlike the pristine organization of the early embryo, differentiation in embryoid bodies appears to be largely unpatterned, although multiple cell types form. It has recently been possible to separate the desired cell types from differentiating ES cells in embryoid bodies by using cell-type-restricted promoters driving expression of either antibiotic resistance genes or fluorophores such as EGFP. In combination with growth factor exposure, highly differentiated cell types have successfully been derived from ES cells. Recent technological advances such as RNA interference to knock down gene expression in ES cells are also producing enriched populations of cells and elucidating gene function in early development.
Collapse
Affiliation(s)
- K Sue O'Shea
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0616, USA.
| |
Collapse
|
39
|
Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 2003; 113:643-55. [PMID: 12787505 DOI: 10.1016/s0092-8674(03)00392-1] [Citation(s) in RCA: 2386] [Impact Index Per Article: 108.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Embryonic stem (ES) cells undergo extended proliferation while remaining poised for multilineage differentiation. A unique network of transcription factors may characterize self-renewal and simultaneously suppress differentiation. We applied expression cloning in mouse ES cells to isolate a self-renewal determinant. Nanog is a divergent homeodomain protein that directs propagation of undifferentiated ES cells. Nanog mRNA is present in pluripotent mouse and human cell lines, and absent from differentiated cells. In preimplantation embryos, Nanog is restricted to founder cells from which ES cells can be derived. Endogenous Nanog acts in parallel with cytokine stimulation of Stat3 to drive ES cell self-renewal. Elevated Nanog expression from transgene constructs is sufficient for clonal expansion of ES cells, bypassing Stat3 and maintaining Oct4 levels. Cytokine dependence, multilineage differentiation, and embryo colonization capacity are fully restored upon transgene excision. These findings establish a central role for Nanog in the transcription factor hierarchy that defines ES cell identity.
Collapse
Affiliation(s)
- Ian Chambers
- Institute for Stem Cell Research, University of Edinburgh, King's Buildings, West Mains Road, Edinburgh EH9 3JQ, Scotland.
| | | | | | | | | | | | | |
Collapse
|
40
|
Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, Maruyama M, Maeda M, Yamanaka S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 2003; 113:631-42. [PMID: 12787504 DOI: 10.1016/s0092-8674(03)00393-3] [Citation(s) in RCA: 2326] [Impact Index Per Article: 105.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Embryonic stem (ES) cells derived from the inner cell mass (ICM) of blastocysts grow infinitely while maintaining pluripotency. Leukemia inhibitory factor (LIF) can maintain self-renewal of mouse ES cells through activation of Stat3. However, LIF/Stat3 is dispensable for maintenance of ICM and human ES cells, suggesting that the pathway is not fundamental for pluripotency. In search of a critical factor(s) that underlies pluripotency in both ICM and ES cells, we performed in silico differential display and identified several genes specifically expressed in mouse ES cells and preimplantation embryos. We found that one of them, encoding the homeoprotein Nanog, was capable of maintaining ES cell self-renewal independently of LIF/Stat3. nanog-deficient ICM failed to generate epiblast and only produced parietal endoderm-like cells. nanog-deficient ES cells lost pluripotency and differentiated into extraembryonic endoderm lineage. These data demonstrate that Nanog is a critical factor underlying pluripotency in both ICM and ES cells.
Collapse
Affiliation(s)
- Kaoru Mitsui
- Laboratory of Animal Molecular Technology, Research and Education Center for Genetic Information, Nara Institute of Science and Technology, Nara 630-0192, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Takahashi K, Mitsui K, Yamanaka S. Role of ERas in promoting tumour-like properties in mouse embryonic stem cells. Nature 2003; 423:541-5. [PMID: 12774123 DOI: 10.1038/nature01646] [Citation(s) in RCA: 257] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2003] [Accepted: 04/01/2003] [Indexed: 01/09/2023]
Abstract
Embryonic stem (ES) cells are pluripotent cells derived from early mammalian embryos. Their immortality and rapid growth make them attractive sources for stem cell therapies; however, they produce tumours (teratomas) when transplanted, which could preclude their therapeutic usage. Why ES cells, which lack chromosomal abnormalities, possess tumour-like properties is largely unknown. Here we show that mouse ES cells specifically express a Ras-like gene, which we have named ERas. We show that human HRasp, which is a recognized pseudogene, does not contain reported base substitutions and instead encodes the human orthologue of ERas. This protein contains amino-acid residues identical to those present in active mutants of Ras and causes oncogenic transformation in NIH 3T3 cells. ERas interacts with phosphatidylinositol-3-OH kinase but not with Raf. ERas-null ES cells maintain pluripotency but show significantly reduced growth and tumorigenicity, which are rescued by expression of ERas complementary DNA or by activated phosphatidylinositol-3-OH kinase. We conclude that the transforming oncogene ERas is important in the tumour-like growth properties of ES cells.
Collapse
Affiliation(s)
- Kazutoshi Takahashi
- Laboratory of Animal Molecular Technology, Research and Education Center for Genetic Information, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | | | | |
Collapse
|
42
|
Abstract
Development of replacement cell therapies awaits the identification of factors that regulate nuclear reprogramming and the mechanisms that control stem cell renewal and differentiation. Once such factors and signals will begin to be elucidated, new technologies will have to be envisaged where uniform differentiation of adult or embryonic stem cells along one differentiation pathway can be induced. Controlled differentiation of stem cells will require the engineering of niches and extracellular signal combinations that would amplify a particular signaling network and allow uniform and selective differentiation. Three recent advances in stem cell research open the possibility to approach engineering studies for cell replacement therapies. Fusion events between stem cells and adult cells or between adult and embryonic stem cells have been shown to result in altered fates and nuclear reprogramming of cell hybrids. Hematopoietic stem cells were shown to require Wnt signaling in order to renew. The purification of Wnt proteins would allow their use as exogenous purified cytokines in attempts to amplify stem cells before bone marrow transplantation. The homeodomain protein Nanog has been shown to be crucial for the embryonic stem cell renewal and pluripotency. However, the cardinal question of how stemness is preserved in the early embryo and adult stem cells remains opened.
Collapse
Affiliation(s)
- S Constantinescu
- Signal Transduction Unit, Ludwig Institute for Cancer Research, Christian de Duve Institute of Cellular Pathology, UCL, Avenue Hippocrate 74, UCL 74+4, Brussels B-1200, Belgium.
| |
Collapse
|
43
|
Tokuzawa Y, Kaiho E, Maruyama M, Takahashi K, Mitsui K, Maeda M, Niwa H, Yamanaka S. Fbx15 is a novel target of Oct3/4 but is dispensable for embryonic stem cell self-renewal and mouse development. Mol Cell Biol 2003; 23:2699-708. [PMID: 12665572 PMCID: PMC152544 DOI: 10.1128/mcb.23.8.2699-2708.2003] [Citation(s) in RCA: 198] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Embryonic stem (ES) cells are immortal and pluripotent cells derived from early mammalian embryos. Transcription factor Oct3/4 is essential for self-renewal of ES cells and early mouse development. However, only a few Oct3/4 target genes have been identified. In this study, we found that F-box-containing protein Fbx15 was expressed predominantly in mouse undifferentiated ES cells. Inactivation of Oct3/4 in ES cells led to rapid extinction of Fbx15 expression. Reporter gene analyses demonstrated that this ES cell-specific expression required an 18-bp enhancer element located approximately 500 nucleotides upstream from the transcription initiation site. The enhancer contained an octamer-like motif and an adjacent Sox-binding motif. Deletion or point mutation of either motif abolished the enhancer activity. The 18-bp fragment became active in NIH 3T3 cells when Oct3/4 and Sox2 were coexpressed. A gel mobility shift assay demonstrated cooperative binding of Oct3/4 and Sox2 to the enhancer sequence. In mice having a beta-galactosidase gene knocked into the Fbx15 locus, 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside staining was detected in ES cells, early embryos (two-cell to blastocyst stages), and testis tissue. Despite such specific expression of Fbx15, homozygous mutant mice showed no gross developmental defects and were fertile. Fbx15-null ES cells were normal in morphology, proliferation, and differentiation. These data demonstrate that Fbx15 is a novel target of Oct3/4 but is dispensable for ES cell self-renewal, development, and fertility.
Collapse
Affiliation(s)
- Yoshimi Tokuzawa
- Laboratory of Animal Molecular Technology, Research and Education Center for Genetic Information, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Nakagawa T, Abe M, Yamazaki T, Miyashita H, Niwa H, Kokubun S, Sato Y. HEX acts as a negative regulator of angiogenesis by modulating the expression of angiogenesis-related gene in endothelial cells in vitro. Arterioscler Thromb Vasc Biol 2003; 23:231-7. [PMID: 12588764 DOI: 10.1161/01.atv.0000052670.55321.87] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The hematopoietically expressed homeobox (HEX) is transiently expressed in endothelial cells (ECs) during vascular formation in embryo. Here, we investigated whether HEX played any role in angiogenesis-related properties of ECs in vitro. METHODS AND RESULTS We transiently overexpressed HEX in human umbilical vein ECs (HUVECs). To our surprise, HEX completely abrogated the response of HUVECs to vascular endothelial growth factor (VEGF) with regard to proliferation, migration, and invasion and abolished network formation by HUVECs on Matrigel. cDNA microarray analysis and quantitative real-time reverse transcription-polymerase chain reaction combined with Western blotting revealed that HEX significantly repressed the expression of VEGF receptor-1, VEGF receptor-2, neuropilin-1, tyrosine kinase with Ig and EGF homology domains (TIE)-1, TIE-2, and the integrin alpha(v) subunit, whereas it augmented the expression of endoglin in HUVECs. We established murine embryonic stem cells that were stably transfected with HEX sense cDNA or antisense cDNA, and we examined the in vitro differentiation to ECs. Although the expression of VEGF receptor-2 was decreased in sense transfectants, the number of cells expressing VE-cadherin, a specific marker of ECs, was not altered. CONCLUSIONS Our present results suggest that HEX may not affect the differentiation of ECs but acts as a negative regulator of angiogenesis.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Differentiation/genetics
- Cell Differentiation/physiology
- Cell Line
- Cell Movement/genetics
- Cells, Cultured
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Developmental/physiology
- Genes/genetics
- Genes, Homeobox/physiology
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/physiology
- Humans
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice
- Neovascularization, Physiologic/genetics
- Neovascularization, Physiologic/physiology
- Oligonucleotide Array Sequence Analysis
- RNA, Messenger/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction
- Stem Cells/chemistry
- Stem Cells/metabolism
- Stem Cells/physiology
- Transcription Factors/genetics
- Transcription Factors/physiology
- Umbilical Veins/chemistry
- Umbilical Veins/cytology
- Umbilical Veins/metabolism
Collapse
Affiliation(s)
- Tomowaki Nakagawa
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Aubert J, Dunstan H, Chambers I, Smith A. Functional gene screening in embryonic stem cells implicates Wnt antagonism in neural differentiation. Nat Biotechnol 2002; 20:1240-5. [PMID: 12447396 DOI: 10.1038/nbt763] [Citation(s) in RCA: 236] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2002] [Accepted: 10/08/2002] [Indexed: 12/22/2022]
Abstract
The multilineage differentiation capacity of mouse embryonic stem (ES) cells offers a potential testing platform for gene products that mediate mammalian lineage determination and cellular specialization. Identification of such differentiation regulators is crucial to harnessing ES cells for pharmaceutical discovery and cell therapy. Here we describe the use of episomal expression technology for functional evaluation of cDNA clones during ES-cell differentiation in vitro. Several candidate cDNAs identified by subtractive cloning and expression profiling were introduced into ES cells in episomal expression constructs. Subsequent differentiation revealed that the Wnt antagonist Sfrp2 stimulates production of neural progenitors. The significance of this observation was substantiated by forced expression of Wnt-1 and treatment with lithium chloride, both of which inhibit neural differentiation. These findings reveal the importance of Wnt signaling in regulating ES-cell lineage diversification. More generally, this study establishes a path for rapid and direct validation of candidate genes in ES cells.
Collapse
Affiliation(s)
- Jerôme Aubert
- Institute for Stem Cell Research, University of Edinburgh, The King's Buildings, West Mains Road, Edinburgh, UK EH3 9JQ
| | | | | | | |
Collapse
|
46
|
Jackson M, Baird JW, Cambray N, Ansell JD, Forrester LM, Graham GJ. Cloning and characterization of Ehox, a novel homeobox gene essential for embryonic stem cell differentiation. J Biol Chem 2002; 277:38683-92. [PMID: 12087094 DOI: 10.1074/jbc.m203459200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We report here the identification and characterization of a novel paired-like homeobox-containing gene (Ehox). This gene, identified in embryonic stem (ES) cells, is differentially expressed during in vitro ES cell differentiation. We have assessed Ehox function using the ES cell in vitro differentiation system. This has involved molecular and biological analyses of the effects of sense or antisense Ehox expression (using episomal vectors) on ES cell differentiation. Analysis of antisense Ehox-expressing ES cells indicates that they are unable to express marker genes associated with hematopoietic, endothelial, or cardiac differentiation following removal of leukemia inhibitory factor. In contrast, overexpression of Ehox using the sense construct accelerated the appearance of these differentiation markers. ES cell self-renewal and differentiation assays reveal that inhibition of Ehox activity results in the maintenance of a stem cell phenotype in limiting concentrations of leukemia inhibitory factor and the almost complete impairment of the cardiomyocyte differentiation capacity of these cells. We therefore conclude that Ehox is a novel homeobox-containing gene that is essential for the earliest stages of murine ES cell differentiation.
Collapse
Affiliation(s)
- Melany Jackson
- John Hughes Bennett Laboratories, Department of Oncology, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Scotland, United Kingdom
| | | | | | | | | | | |
Collapse
|
47
|
Fujikura J, Yamato E, Yonemura S, Hosoda K, Masui S, Nakao K, Miyazaki Ji JI, Niwa H. Differentiation of embryonic stem cells is induced by GATA factors. Genes Dev 2002; 16:784-9. [PMID: 11937486 PMCID: PMC186328 DOI: 10.1101/gad.968802] [Citation(s) in RCA: 395] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Extraembryonic endoderm (ExE) is differentiated from the inner cell mass of the late blastocyst-stage embryo to form visceral and parietal endoderm, both of which have an important role in early embryogenesis. The essential roles of Gata-6 and Gata-4 on differentiation of visceral endoderm have been identified by analyses of knockout mice. Here we report that forced expression of either Gata-6 or Gata-4 in embryonic stem (ES) cells is sufficient to induce the proper differentiation program towards ExE. We believe that this is the first report of a physiological differentiation event induced by the ectopic expression of a transcription factor in ES cells.
Collapse
Affiliation(s)
- Junji Fujikura
- Stem Cell Regulation Research, Area of Molecular Therapeutics, Course of Advanced Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Niwa H, Masui S, Chambers I, Smith AG, Miyazaki JI. Phenotypic complementation establishes requirements for specific POU domain and generic transactivation function of Oct-3/4 in embryonic stem cells. Mol Cell Biol 2002; 22:1526-36. [PMID: 11839818 PMCID: PMC134688 DOI: 10.1128/mcb.22.5.1526-1536.2002] [Citation(s) in RCA: 238] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transcription factors of the POU family govern cell fate through combinatorial interactions with coactivators and corepressors. The POU factor Oct-3/4 can define differentiation, dedifferentation, or self-renewal of pluripotent embryonic stem (ES) cells in a sensitive, dose-dependent manner (H. Niwa, J.-I. Miyazali, and A. G. Smith, Nat. Genet. 24:372-376, 2000). Here we have developed a complementation assay based on the ability of Oct-3/4 transgenes to rescue self-renewal in conditionally null ES cells and used this to define which domains of Oct-3/4 are required to sustain the undifferentiated stem cell phenotype. Surprisingly, we found that molecules lacking either the N-terminal or C-terminal transactivation domain, though not both, can effectively replace full-length Oct-3/4. Furthermore, a fusion of the heterologous transactivation domain of Oct-2 to the Oct-3/4 POU domain can also sustain self-renewal. Thus, the unique function of Oct-3/4 in ES cell propagation resides in combination of the specific POU domain with a generic proline-rich transactivation domain. Interestingly, however, Oct-3/4 target gene expression elicited by the N- and C-terminal transactivation domains is not identical, indicating that at least one class of genes activated by Oct-3/4 is not required for ES cell propagation.
Collapse
Affiliation(s)
- Hitoshi Niwa
- Stem Cell Regulation Research, Area of Molecular Therapeutics, Course of Advanced Medicine, Osaka University Graduate School of Medicine, Suita C, Osaka 565-0871, Japan.
| | | | | | | | | |
Collapse
|
49
|
Hayashi H, Tominaga Y, Hirano S, McKenna AE, Nakabeppu Y, Matsumoto Y. Replication-associated repair of adenine:8-oxoguanine mispairs by MYH. Curr Biol 2002; 12:335-9. [PMID: 11864576 DOI: 10.1016/s0960-9822(02)00686-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Cellular DNA is constantly exposed to the risk of oxidation. 8-oxoguanine (8-oxoG) is one of the major DNA lesions generated by oxidation, which is primarily corrected by base excision repair. When it is not repaired prior to replication, replicative DNA polymerases yield misinsertion of an adenine (A) opposite the 8-oxoG on the template strand, generating an A:8-oxoG mispair. MYH, a mammalian homolog of Escherichia coli MutY, is a DNA glycosylase responsible for initiating base excision repair of such a mispair by excising the adenine opposite 8-oxoG. Here, using an in vivo repair system, we show that DNA replication enhances the repair of the A:8-oxoG mispair. Repair efficiency was lower in MYH-deficient murine cells than in MYH-proficient cells. Transfection of the MYH-deficient cells with a wild-type MYH expression vector increased the efficiency of A:8-oxoG repair, indicating that a significant part of this replication-associated repair depends on MYH. Expression of a mutant MYH in which the PCNA binding motif was disrupted did not increase the repair efficiency, thus suggesting that the interaction between PCNA and MYH is critical for MYH-initiated repair of A:8-oxoG.
Collapse
Affiliation(s)
- Harutoshi Hayashi
- Division of Medical Science, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Mouse embryonic stem cells are continuous cell lines derived directly from the fetal founder tissue of the preimplantation embryo. They can be expanded in culture while retaining the functional attributes of pluripotent early embryo cells. In particular, they can participate fully in fetal development when reintroduced into the embryo. The capacity for multilineage differentiation is reproduced in culture where embryonic stem cells can produce a wide range of well-defined cell types. This has stimulated interest in the isolation of analogous cells of human origin. Such human pluripotent stem cells could constitute a renewable source of more differentiated cells that could be employed to replace diseased or damaged tissue by cellular transplantation. In this review, the relationships between mouse embryonic stem cells, resident pluripotent cells in the embryo, and human embryo-derived cell lines are evaluated, and the prospects and challenges of embryo stem cell research are considered.
Collapse
Affiliation(s)
- A G Smith
- Centre for Genome Research, University of Edinburgh, The King's Buildings, West Mains Road, Edinburgh, Scotland, EH9 3JQ, United Kingdom.
| |
Collapse
|