251
|
Cova E, Ghiroldi A, Guareschi S, Mazzini G, Gagliardi S, Davin A, Bianchi M, Ceroni M, Cereda C. G93A SOD1 alters cell cycle in a cellular model of Amyotrophic Lateral Sclerosis. Cell Signal 2010; 22:1477-84. [PMID: 20561900 DOI: 10.1016/j.cellsig.2010.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 05/26/2010] [Indexed: 12/14/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative multifactorial disease characterized, like other diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) or frontotemporal dementia (FTD), by the degeneration of specific neuronal cell populations. Motor neuron loss is distinctive of ALS. However, the causes of onset and progression of motor neuron death are still largely unknown. In about 2% of all cases, mutations in the gene encoding for the Cu/Zn superoxide dismutase (SOD1) are implicated in the disease. Several alterations in the expression or activation of cell cycle proteins have been described in the neurodegenerative diseases and related to cell death. In this work we show that mutant SOD1 can alter cell cycle in a cellular model of ALS. Our findings suggest that modifications in the cell cycle progression could be due to an increased interaction between mutant G93A SOD1 and Bcl-2 through the cyclins regulator p27. As previously described in post mitotic neurons, cell cycle alterations could fatally lead to cell death.
Collapse
Affiliation(s)
- Emanuela Cova
- Laboratory of Experimental Neurobiology, IRCCS, National Neurological Institute C. Mondino, Via Mondino, 2, 27100 Pavia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
252
|
Rodin S, Domogatskaya A, Ström S, Hansson EM, Chien KR, Inzunza J, Hovatta O, Tryggvason K. Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511. Nat Biotechnol 2010; 28:611-5. [PMID: 20512123 DOI: 10.1038/nbt.1620] [Citation(s) in RCA: 399] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 03/08/2010] [Indexed: 12/11/2022]
Abstract
We describe a system for culturing human embryonic stem (hES) cells and induced pluripotent stem (iPS) cells on a recombinant form of human laminin-511, a component of the natural hES cell niche. The system is devoid of animal products and feeder cells and contains only one undefined component, human albumin. The hES cells self-renewed with normal karyotype for at least 4 months (20 passages), after which the cells could produce teratomas containing cell lineages of all three germ layers. When plated on laminin-511 in small clumps, hES cells spread out in a monolayer, maintaining cellular homogeneity with approximately 97% OCT4-positive cells. Adhesion of hES cells was dependent on alpha6beta1 integrin. The use of homogeneous monolayer hES or iPS cell cultures provides more controllable conditions for the design of differentiation methods. This xeno-free and feeder-free system may be useful for the development of cell lineages for therapeutic purposes.
Collapse
Affiliation(s)
- Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
253
|
Iacobas I, Vats A, Hirschi KK. Vascular potential of human pluripotent stem cells. Arterioscler Thromb Vasc Biol 2010; 30:1110-7. [PMID: 20453170 DOI: 10.1161/atvbaha.109.191601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is the number one cause of death and disability in the US. Understanding the biological activity of stem and progenitor cells, and their ability to contribute to the repair, regeneration and remodeling of the heart and blood vessels affected by pathological processes is an essential part of the paradigm in enabling us to achieve a reduction in related deaths. Both human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells are promising sources of cells for clinical cardiovascular therapies. Additional in vitro studies are needed, however, to understand their relative phenotypes and molecular regulation toward cardiovascular cell fates. Further studies in translational animal models are also needed to gain insights into the potential and function of both human ES- and iPS-derived cardiovascular cells, and enable translation from experimental and preclinical studies to human trials.
Collapse
Affiliation(s)
- Ionela Iacobas
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | |
Collapse
|
254
|
Li W, Liu C, Qin J, Zhang L, Chen R, Chen J, Yu X, Wu G, Lahn BT, Fu Y, Xiang AP. Efficient genetic modification of cynomolgus monkey embryonic stem cells with lentiviral vectors. Cell Transplant 2010; 19:1181-93. [PMID: 20447344 DOI: 10.3727/096368910x504469] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Embryonic stem (ES) cells have the ability to undergo indefinite self-renewal in vitro and give rise during development to derivatives of all three primary germ layers (ectoderm, endoderm, and mesoderm), which make them a highly prized reagent in cell and gene therapy. Efficient introduction of various genes of interest into primate ES cells has proven to be difficult. Here, we demonstrated that the self-inactivating HIV-1-based lentiviral vectors constructed by MultiSite gateway technology are efficient tools for the transduction of cynomolgus monkey (Macaca fasicularis) ES (cmES) cells. After antibiotic selection, all of the transduced cells can stably express the reporter gene (humanized Renilla GFP or dTomato) while maintaining their stem cell properties, including continuous expression of stem cell markers, alkaline phosphatase (AKP), OCT-4, SSEA-4, and TRA-1-60, formation of embryoid bodies in vitro and teratomas in vivo containing derivatives of three embryonic germ layers. This approach will provide a useful tool for both gene function studies and in vivo cell tracking of stem cells.
Collapse
Affiliation(s)
- Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
255
|
Fernandes AM, Meletti T, Guimarães R, Stelling MP, Marinho PAN, Valladão AS, Rehen SK. Worldwide Survey of Published Procedures to Culture Human Embryonic Stem Cells. Cell Transplant 2010; 19:509-23. [DOI: 10.3727/096368909x485067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Since their derivation 11 years ago, human embryonic stem (hES) cells have become a powerful tool in both basic biomedical research and developmental biology. Their capacity for self-renewal and differentiation into any tissue type has also brought interest from fields such as cell therapy and drug screening. We conducted an extensive analysis of 750 papers (51% of the total published about hES cells between 1998 and 2008) to present a spectrum of hES cell research including culture protocols developed worldwide. This review may stimulate discussions about the importance of having unvarying methods to culture hES cells, in order to facilitate comparisons among data obtained by research groups elsewhere, especially concerning preclinical studies. Moreover, the description of the most widely used cell lines, reagents, and procedures adopted internationally will help newcomers on deciding the best strategies for starting their own studies. Finally, the results will contribute with the efforts of stem cell researchers on comparing the performance of different aspects related to hES cell culture methods.
Collapse
Affiliation(s)
- A. M. Fernandes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - T. Meletti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - R. Guimarães
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - M. P. Stelling
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P. A. N. Marinho
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Engenharia Química/COPPE, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - A. S. Valladão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - S. K. Rehen
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
256
|
Galvin KE, Travis ED, Yee D, Magnuson T, Vivian JL. Nodal signaling regulates the bone morphogenic protein pluripotency pathway in mouse embryonic stem cells. J Biol Chem 2010; 285:19747-56. [PMID: 20427282 DOI: 10.1074/jbc.m109.077347] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Members of the transforming growth factor-beta superfamily play essential roles in both the pluripotency and differentiation of embryonic stem (ES) cells. Although bone morphogenic proteins (BMPs) maintain pluripotency of undifferentiated mouse ES cells, the role of autocrine Nodal signaling is less clear. Pharmacological, molecular, and genetic methods were used to further understand the roles and potential interactions of these pathways. Treatment of undifferentiated ES cells with SB431542, a pharmacological inhibitor of Smad2 signaling, resulted in a rapid reduction of phosphorylated Smad2 and altered the expression of several putative downstream targets. Unexpectedly, inhibition of the Nodal signaling pathway resulted in enhanced BMP signaling, as assessed by Smad1/5 phosphorylation. SB431542-treated cells also demonstrated significant induction of the Id genes, which are known direct targets of BMP signaling and important factors in ES cell pluripotency. Inhibition of BMP signaling decreased the SB431542-mediated phosphorylation of Smad1/5 and induction of Id genes, suggesting that BMP signaling is necessary for some Smad2-mediated activity. Because Smad7, a known inhibitory factor to both Nodal and BMP signaling, was down-regulated following inhibition of Nodal-Smad2 signaling, the contribution of Smad7 to the cross-talk between the transforming growth factor-beta pathways in ES cells was examined. Biochemical manipulation of Smad7 expression, through shRNA knockdown or inducible gene expression, significantly reduced the SB431542-mediated phosphorylation of Smad1/5 and induction of the Id genes. We conclude that autocrine Nodal signaling in undifferentiated mouse ES cells modulates the vital pluripotency pathway of BMP signaling.
Collapse
Affiliation(s)
- Katherine E Galvin
- Department of Pathology and Laboratory Medicine and Division of Cancer and Developmental Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | |
Collapse
|
257
|
Culture Conditions and Signalling Networks Promoting the Establishment of Cell Lines from Parthenogenetic and Biparental Pig Embryos. Stem Cell Rev Rep 2010; 6:484-95. [DOI: 10.1007/s12015-010-9153-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
258
|
Cai J, Xie D, Fan Z, Chipperfield H, Marden J, Wong WH, Zhong S. Modeling co-expression across species for complex traits: insights to the difference of human and mouse embryonic stem cells. PLoS Comput Biol 2010; 6:e1000707. [PMID: 20300647 PMCID: PMC2837392 DOI: 10.1371/journal.pcbi.1000707] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 02/05/2010] [Indexed: 01/14/2023] Open
Abstract
Complex interactions between genes or proteins contribute substantially to phenotypic evolution. We present a probabilistic model and a maximum likelihood approach for cross-species clustering analysis and for identification of conserved as well as species-specific co-expression modules. This model enables a “soft” cross-species clustering (SCSC) approach by encouraging but not enforcing orthologous genes to be grouped into the same cluster. SCSC is therefore robust to obscure orthologous relationships and can reflect different functional roles of orthologous genes in different species. We generated a time-course gene expression dataset for differentiating mouse embryonic stem (ES) cells, and compiled a dataset of published gene expression data on differentiating human ES cells. Applying SCSC to analyze these datasets, we identified conserved and species-specific gene regulatory modules. Together with protein-DNA binding data, an SCSC cluster specifically induced in murine ES cells indicated that the KLF2/4/5 transcription factors, although critical to maintaining the pluripotent phenotype in mouse ES cells, were decoupled from the OCT4/SOX2/NANOG regulatory module in human ES cells. Two of the target genes of murine KLF2/4/5, LIN28 and NODAL, were rewired to be targets of OCT4/SOX2/NANOG in human ES cells. Moreover, by mapping SCSC clusters onto KEGG signaling pathways, we identified the signal transduction components that were induced in pluripotent ES cells in either a conserved or a species-specific manner. These results suggest that the pluripotent cell identity can be established and maintained through more than one gene regulatory network. A major goal in biology is to understand the evolution of complex traits, such as the development of multicellular body plans. To a certain extent, complex traits are governed by regulated gene expression. The comparison expression data between species requires extra considerations than sequence comparison, because gene expression is not static and the level of expression is influenced by external conditions. Considering that co-expression patterns are often comparable across species, we developed a statistical model for cross-species clustering analysis. The model allows each species to create its own clusters of the genes but also encourages the species to borrow strength from each others' clusters of orthologous genes. The result is a pairing of clusters, one from each species, where the paired clusters share many but not necessarily all orthologous genes. The model-based approach not only reduces subjective influence but also enables effective use of evolutionary dependence. Applying this model to analyze human and mouse embryonic stem (ES) cell data, we identified the transcription factors and the signaling proteins that are specifically expressed in either human or mouse ES cells. These results suggest that the pluripotent cell identity can be established and maintained through more than one gene regulatory network.
Collapse
Affiliation(s)
- Jun Cai
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
| | - Dan Xie
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
| | - Zhewen Fan
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
- Department of Statistics, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
| | | | - John Marden
- Department of Statistics, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
| | - Wing H. Wong
- Department of Statistics, Stanford University, Stanford, California, United States of America
- Department of Health Research and Policy, Stanford University, Stanford, California, United States of America
| | - Sheng Zhong
- Department of Bioengineering, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
- Department of Statistics, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
- Institute of Genomic Biology, University of Illinois at Urbana Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
259
|
Xie D, Chen CC, Ptaszek LM, Xiao S, Cao X, Fang F, Ng HH, Lewin HA, Cowan C, Zhong S. Rewirable gene regulatory networks in the preimplantation embryonic development of three mammalian species. Genome Res 2010; 20:804-15. [PMID: 20219939 DOI: 10.1101/gr.100594.109] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Mammalian preimplantation embryonic development (PED) is thought to be governed by highly conserved processes. While it had been suggested that some plasticity of conserved signaling networks exists among different mammalian species, it was not known to what extent modulation of the genomes and the regulatory proteins could "rewire" the gene regulatory networks (GRN) that control PED. We therefore generated global transcriptional profiles from three mammalian species (human, mouse, and bovine) at representative stages of PED, including: zygote, two-cell, four-cell, eight-cell, 16-cell, morula and blastocyst. Coexpression network analysis suggested that 40.2% orthologous gene triplets exhibited different expression patterns among these species. Combining the expression data with genomic sequences and the ChIP-seq data of 16 transcription regulators, we observed two classes of genomic changes that contributed to interspecies expression difference, including single nucleotide mutations leading to turnover of transcription factor binding sites, and insertion of cis-regulatory modules (CRMs) by transposons. About 10% of transposons are estimated to carry CRMs, which may drive species-specific gene expression. The two classes of genomic changes act in concert to drive mouse-specific expression of MTF2, which links POU5F1/NANOG to NOTCH signaling. We reconstructed the transition of the GRN structures as a function of time during PED. A comparison of the GRN transition processes among the three species suggested that in the bovine system, POU5F1's interacting partner SOX2 may be replaced by HMGB1 (a TF sharing the same DNA binding domain with SOX2), resulting in rewiring of GRN by a trans change.
Collapse
Affiliation(s)
- Dan Xie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
260
|
Sarkadi B, Orbán TI, Szakács G, Várady G, Schamberger A, Erdei Z, Szebényi K, Homolya L, Apáti A. Evaluation of ABCG2 expression in human embryonic stem cells: crossing the same river twice? Stem Cells 2010; 28:174-6. [PMID: 19924769 DOI: 10.1002/stem.262] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
261
|
Shimozawa N, Nakamura S, Takahashi I, Hatori M, Sankai T. Characterization of a novel embryonic stem cell line from an ICSI-derived blastocyst in the African green monkey. Reproduction 2010; 139:565-73. [DOI: 10.1530/rep-09-0067] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Several cell types from the African green monkey (Cercopithecus aethiops), such as red blood cells, primary culture cells from kidney, and the Vero cell line, are valuable sources for biomedical research and testing. Embryonic stem (ES) cells that are established from blastocysts have pluripotency to differentiate into these and other types of cells. We examined an in vitro culture system of zygotes produced by ICSI in African green monkeys and attempted to establish ES cells. Culturing with and without a mouse embryonic fibroblast (MEF) cell monolayer resulted in the development of ICSI-derived zygotes to the blastocyst stage, while culturing with a buffalo rat liver cell monolayer yielded no development (3/14, 21.4% and 6/31, 19.4% vs 0/23, 0% respectively; P<0.05). One of the nine blastocysts, which had been one of the zygotes co-cultured with MEF cells, formed flat colonies consisting of cells with large nuclei, similar to other primate ES cell lines. The African green monkey ES (AgMES) cells expressed pluripotency markers, formed teratomas consisting of three embryonic germ layer tissues, and had a normal chromosome number. Furthermore, expression of the germ cell markers CD9 and DPPA3 (STELLA) was detected in the embryoid bodies, suggesting that AgMES cells might have the potential ability to differentiate into germ cells. The results suggested that MEF cells greatly affected the quality of the inner cell mass of the blastocysts. In addition, AgMES cells would be a precious resource for biomedical research such as other primate ES cell lines.
Collapse
|
262
|
Markovà E, Malmgren LO, Belyaev IY. Microwaves from Mobile Phones Inhibit 53BP1 Focus Formation in Human Stem Cells More Strongly Than in Differentiated Cells: Possible Mechanistic Link to Cancer Risk. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:394-399. [PMID: 20064781 PMCID: PMC2854769 DOI: 10.1289/ehp.0900781] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 10/22/2009] [Indexed: 05/26/2023]
Abstract
BACKGROUND It is widely accepted that DNA double-strand breaks (DSBs) and their misrepair in stem cells are critical events in the multistage origination of various leukemias and tumors, including gliomas. OBJECTIVES We studied whether microwaves from mobile telephones of the Global System for Mobile Communication (GSM) and the Universal Global Telecommunications System (UMTS) induce DSBs or affect DSB repair in stem cells. METHODS We analyzed tumor suppressor TP53 binding protein 1 (53BP1) foci that are typically formed at the sites of DSB location (referred to as DNA repair foci) by laser confocal microscopy. RESULTS Microwaves from mobile phones inhibited formation of 53BP1 foci in human primary fibroblasts and mesenchymal stem cells. These data parallel our previous findings for human lymphocytes. Importantly, the same GSM carrier frequency (915 MHz) and UMTS frequency band (1947.4 MHz) were effective for all cell types. Exposure at 905 MHz did not inhibit 53BP1 foci in differentiated cells, either fibroblasts or lymphocytes, whereas some effects were seen in stem cells at 905 MHz. Contrary to fibroblasts, stem cells did not adapt to chronic exposure during 2 weeks. CONCLUSIONS The strongest microwave effects were always observed in stem cells. This result may suggest both significant misbalance in DSB repair and severe stress response. Our findings that stem cells are most sensitive to microwave exposure and react to more frequencies than do differentiated cells may be important for cancer risk assessment and indicate that stem cells are the most relevant cellular model for validating safe mobile communication signals.
Collapse
Affiliation(s)
- Eva Markovà
- Department of Genetics, Microbiology and Toxicology, Stockholm University, Stockholm, Sweden
- Laboratory of Molecular Genetics, Cancer Research Institute, Bratislava, Slovak Republic
| | | | - Igor Y. Belyaev
- Department of Genetics, Microbiology and Toxicology, Stockholm University, Stockholm, Sweden
- Laboratory of Molecular Genetics, Cancer Research Institute, Bratislava, Slovak Republic
- Laboratory of Radiobiology, General Physics Institute, Russian Academy of Science, Moscow, Russia
| |
Collapse
|
263
|
Akopian V, Andrews PW, Beil S, Benvenisty N, Brehm J, Christie M, Ford A, Fox V, Gokhale PJ, Healy L, Holm F, Hovatta O, Knowles BB, Ludwig TE, McKay RDG, Miyazaki T, Nakatsuji N, Oh SKW, Pera MF, Rossant J, Stacey GN, Suemori H. Comparison of defined culture systems for feeder cell free propagation of human embryonic stem cells. In Vitro Cell Dev Biol Anim 2010; 46:247-58. [PMID: 20186512 PMCID: PMC2855804 DOI: 10.1007/s11626-010-9297-z] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 01/15/2010] [Indexed: 12/23/2022]
Abstract
There are many reports of defined culture systems for the propagation of human embryonic stem cells in the absence of feeder cell support, but no previous study has undertaken a multi-laboratory comparison of these diverse methodologies. In this study, five separate laboratories, each with experience in human embryonic stem cell culture, used a panel of ten embryonic stem cell lines (including WA09 as an index cell line common to all laboratories) to assess eight cell culture methods, with propagation in the presence of Knockout Serum Replacer, FGF-2, and mouse embryonic fibroblast feeder cell layers serving as a positive control. The cultures were assessed for up to ten passages for attachment, death, and differentiated morphology by phase contrast microscopy, for growth by serial cell counts, and for maintenance of stem cell surface marker expression by flow cytometry. Of the eight culture systems, only the control and those based on two commercial media, mTeSR1 and STEMPRO, supported maintenance of most cell lines for ten passages. Cultures grown in the remaining media failed before this point due to lack of attachment, cell death, or overt cell differentiation. Possible explanations for relative success of the commercial formulations in this study, and the lack of success with other formulations from academic groups compared to previously published results, include: the complex combination of growth factors present in the commercial preparations; improved development, manufacture, and quality control in the commercial products; differences in epigenetic adaptation to culture in vitro between different ES cell lines grown in different laboratories.
Collapse
Affiliation(s)
-
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
264
|
Comparative Analysis of Five Different Homologous Feeder Cell Lines in the Ability to Support Rhesus Embryonic Stem Cells. Zool Res 2010. [DOI: 10.3724/sp.j.1141.2009..04345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
265
|
Gerecht S, Ferreira LS, Langer R. Vascular differentiation of human embryonic stem cells in bioactive hydrogel-based scaffolds. Methods Mol Biol 2010; 584:333-54. [PMID: 19907986 DOI: 10.1007/978-1-60761-369-5_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The vascularization of tissue constructs remains a major challenge in regenerative medicine, as the diffusional supply of oxygen can support only 100-200 mum thick layers of viable tissue. The formation of a mature and functional vascular network requires communication between endothelial cells (ECs) and smooth muscle cells (SMCs). Potential sources of these cells that involve noninvasive methodologies are required for numerous applications including tissue-engineered vascular grafts, myocardial ischemia, wound healing, plastic surgery, and general tissue-engineering applications. Human embryonic stem cells (hESCs) can be an unlimited source of these cells. They can be expanded in vitro in an undifferentiated state without apparent limit, and hES-derived cells can be created in virtually unlimited amounts for potential clinical uses. Recently, vascular progenitor cells as well as endothelial and smooth muscle cells have been isolated from hESCs.
Collapse
Affiliation(s)
- Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|
266
|
Huang B, Cui K, Li T, Wang X, Lu F, Liu Q, da Silva FM, Shi D. Generation of Buffalo (Bubalus bubalis) Transgenic Chimeric and Nuclear Transfer Embryos Using Embryonic Germ-Like Cells Expressing Enhanced Green Fluorescent Protein. Reprod Domest Anim 2010; 45:103-8. [DOI: 10.1111/j.1439-0531.2008.01262.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
267
|
Sokolov MV, Panyutin IV, Onyshchenko MI, Panyutin IG, Neumann RD. Expression of pluripotency-associated genes in the surviving fraction of cultured human embryonic stem cells is not significantly affected by ionizing radiation. Gene 2010; 455:8-15. [PMID: 20123005 DOI: 10.1016/j.gene.2010.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 01/08/2010] [Accepted: 01/24/2010] [Indexed: 01/29/2023]
Abstract
Human embryonic stem cells (hESC) are capable to give rise to all cell types in the human body during the normal course of development. Therefore, these cells hold a great promise in regenerative cell replacement based therapeutical approaches. However, some controversy exists in literature concerning the ultimate fate of hESC after exposure to genotoxic agents, in particular, regarding the effect of DNA damaging insults on pluripotency of hESC. To comprehensively address this issue, we performed an analysis of the expression of marker genes, associated with pluripotent state of hESC, such as Oct-4, Nanog, Sox-2, SSEA-4, TERT, TRA-1-60 and TRA-1-81 up to 65h after exposure to ionizing radiation (IR) using flow cytometry, immunocytochemistry and quantitative real-time polymerase chain reaction techniques. We show that irradiation with relatively low doses of gamma-radiation (0.2Gy and 1Gy) does not lead to loss of expression of the pluripotency-associated markers in the surviving hESC. While changes in the levels of expression of some of the pluripotency markers were observed at different time points after IR exposure, these alterations were not persistent, and, in most cases, the expression of the pluripotency-associated markers remained significantly higher than that observed in fully differentiated human fibroblasts, and in hESCs differentiated into definitive endodermal lineage. Our data suggest that exposure of hESC to relatively low doses of IR as a model genotoxic agent does not significantly affect pluripotency of the surviving fraction of hESC.
Collapse
Affiliation(s)
- Mykyta V Sokolov
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
268
|
Huang B, Li T, Wang XL, Xie TS, Lu YQ, da Silva FM, Shi DS. Generation and Characterization of Embryonic Stem-Like Cell Lines Derived fromIn VitroFertilization Buffalo (Bubalus bubalis) Embryos. Reprod Domest Anim 2010; 45:122-8. [DOI: 10.1111/j.1439-0531.2008.01268.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
269
|
Xue H, Wu S, Papadeas ST, Spusta S, Swistowska AM, MacArthur CC, Mattson MP, Maragakis NJ, Capecchi MR, Rao MS, Zeng X, Liu Y. A targeted neuroglial reporter line generated by homologous recombination in human embryonic stem cells. Stem Cells 2010; 27:1836-46. [PMID: 19544414 DOI: 10.1002/stem.129] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this study, we targeted Olig2, a basic helix-loop-helix transcription factor that plays an important role in motoneuron and oligodendrocyte development, in human embryonic stem cell (hESC) line BG01 by homologous recombination. One allele of Olig2 locus was replaced by a green fluorescent protein (GFP) cassette with a targeting efficiency of 5.7%. Targeted clone R-Olig2 (like the other clones) retained pluripotency, typical hESC morphology, and a normal parental karyotype 46,XY. Most importantly, GFP expression recapitulated endogenous Olig2 expression when R-Olig2 was induced by sonic hedgehog and retinoic acid, and GFP-positive cells could be purified by fluorescence-activated cell sorting. Consistent with previous reports on rodents, early GFP-expressing cells appeared biased to a neuronal fate, whereas late GFP-expressing cells appeared biased to an oligodendrocytic fate. This was corroborated by myoblast coculture, transplantation into the rat spinal cords, and whole genome expression profiling. The present work reports an hESC reporter line generated by homologous recombination targeting a neural lineage-specific gene, which can be differentiated and sorted to obtain pure neural progenitor populations.
Collapse
Affiliation(s)
- Haipeng Xue
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California 92008, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
270
|
Barakat TS, Gribnau J. X chromosome inactivation and embryonic stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 695:132-54. [PMID: 21222204 DOI: 10.1007/978-1-4419-7037-4_10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
X chromosome inactivation (XCI) is a process required to equalize the dosage of X-encoded genes between female and male cells. XCI is initiated very early during female embryonic development or upon differentiation of female embryonic stem (ES) cells and results in inactivation of one X chromosome in every female somatic cell. The regulation of XCI involves factors that also play a crucial role in ES cell maintenance and differentiation and the XCI process therefore provides a beautiful paradigm to study ES cell biology. In this chapter we describe the important cis and trans acting regulators of XCI and introduce the models that have been postulated to explain initiation of XCI in female cells only. We also discuss the proteins involved in the establishment of the inactive X chromosome and describe the different chromatin modifications associated with the inactivation process. Finally, we describe the potential of mouse and human ES and induced pluripotent stem (iPS) cells as model systems to study the XCI process.
Collapse
Affiliation(s)
- Tahsin Stefan Barakat
- Department of Reproduction and Development, University Medical Center, Room Ee 09-71, Erasmus MC, 3015 GE, Rotterdam, Netherlands
| | | |
Collapse
|
271
|
Jeong Y, Mangelsdorf DJ. Nuclear receptor regulation of stemness and stem cell differentiation. Exp Mol Med 2009; 41:525-37. [PMID: 19696553 DOI: 10.3858/emm.2009.41.8.091] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stem cells include a diverse number of toti-, pluri-, and multi-potent cells that play important roles in cellular genesis and differentiation, tissue development, and organogenesis. Genetic regulation involving various transcription factors results in the self-renewal and differentiation properties of stem cells. The nuclear receptor (NR) superfamily is composed of 48 ligand-activated transcription factors involved in diverse physiological functions such as metabolism, development, and reproduction. Increasing evidence shows that certain NRs function in regulating stemness or differentiation of embryonic stem (ES) cells and tissue-specific adult stem cells. Here, we review the role of the NR superfamily in various aspects of stem cell biology, including their regulation of stemness, forward- and trans-differentiation events; reprogramming of terminally differentiated cells; and interspecies differences. These studies provide insights into the therapeutic potential of the NR superfamily in stem cell therapy and in treating stem cell-associated diseases (e.g., cancer stem cell).
Collapse
Affiliation(s)
- Yangsik Jeong
- Department of Pharmacology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Texas 75390, USA
| | | |
Collapse
|
272
|
Wwp2 mediates Oct4 ubiquitination and its own auto-ubiquitination in a dosage-dependent manner. Cell Res 2009; 20:332-44. [DOI: 10.1038/cr.2009.136] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
273
|
Schneider MR, Wolf E, Braun J, Kolb HJ, Adler H. Canine embryonic stem cells: state of the art. Theriogenology 2009; 74:492-7. [PMID: 19963261 DOI: 10.1016/j.theriogenology.2009.09.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 09/10/2009] [Accepted: 09/30/2009] [Indexed: 02/07/2023]
Abstract
Embryonic stem cells (ESCs) are permanent cell lines that can be maintained in a pluripotent, undifferentiated state. Appropriate environmental stimuli can cause them to differentiate into cell types of all three germ layers both in vitro and in vivo. Embryonic stem cells bear many opportunities for clinical applications in tissue engineering and regenerative medicine. Whereas most of our knowledge on the biology and technology of ESCs is derived from studies with mouse cells, large animal models mimicking important aspects of human anatomy, physiology, and pathology more closely than mouse models are urgently needed for studies evaluating the safety and efficacy of cell therapies. The dog is an excellent model for studying human diseases, and the availability of canine ESCs would open new possibilities for this model in biomedical research. In addition, canine ESCs could be useful for the development of cell-based approaches for the treatment of dogs. Here, we discuss the features of recently reported canine embryo-derived cells and their potential applications in basic and translational biomedical research.
Collapse
Affiliation(s)
- M R Schneider
- Institut für Molekulare Tierzucht und Biotechnologie, Genzentrum der LMU München, München, Germany.
| | | | | | | | | |
Collapse
|
274
|
Vieyra DS, Rosen A, Goodell MA. Identification and characterization of side population cells in embryonic stem cell cultures. Stem Cells Dev 2009; 18:1155-66. [PMID: 19113897 DOI: 10.1089/scd.2008.0391] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Marker and functional heterogeneity has been described for embryonic stem cells (ESCs). This property has been correlated with the presence of ESC subpopulations resembling pluripotent cell lineages of the embryo. The ability to efflux Hoechst (Ho) displayed by side population (SP) cells has proven valuable as a marker to identify multipotent stem cells from a variety of tissues. Here we report that cultures from different ESC lines consistently show an SP population that displays antigens of undifferentiated ESCs, distinct drug efflux properties, and an expression pattern of ABC transporters, inner cell mass (ICM), and epiblast genes, which distinguish it from the non-SP ESC fraction. This SP population contains pluripotent cells that differentiate into ectoderm, mesoderm, and endoderm in embryoid body and teratoma assays. Further, purified SP cells efficiently integrate into developing morulae and contribute to ICM. Under standard ESC culture conditions, SP and non-SP populations display ability to convert into each other; however, an equilibrium establishes between these fractions. Using protocols customized for SP ESCs, we report that cells with similar efflux properties can be identified in the ICM of peri-implanted blastocysts. Our results indicate that ESCs display heterogeneity for the SP marker, and the SP population of these cultures contains cells that phenotypically and functionally resemble efflux-active ICM cells of the peri-implanted embryo. Our observations suggest an involvement of the SP phenotype in ESC maintenance and early embryo development, and support the idea that ESCs are composed of distinct phenotypic and functional pluripotent subpopulations in dynamic equilibrium.
Collapse
Affiliation(s)
- Diego S Vieyra
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | | | | |
Collapse
|
275
|
Wilcox JT, Semple E, Gartley C, Brisson BA, Perrault SD, Villagómez DAF, Tayade C, Becker S, Lanza R, Betts DH. Characterization of canine embryonic stem cell lines derived from different niche microenvironments. Stem Cells Dev 2009; 18:1167-78. [PMID: 19327015 DOI: 10.1089/scd.2008.0336] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Embryo-derived stem cells hold enormous potential for producing cell-based transplantation therapies, allowing high-throughput drug screening and delineating early embryonic development. However, potential clinical applications must first be tested for safety and efficacy in preclinical animal models. Due to physiological and genetic parity to humans, the domestic dog is widely used as a clinically relevant animal model for cardiovascular, neurodegenerative, orthopedic, and oncologic diseases. Therefore, we established numerous putative canine embryonic stem cell (cESC) lines by immunodissection of the inner cell mass (ICM), which we termed OVC.ID.1-23, and by explant outgrowths from whole canine blastocysts, named OVC.EX.1-16. All characterized lines were immunopositive for OCT4, SOX2, NANOG, SSEA-3, and SSEA-4; displayed high telomerase and alkaline phosphatase (ALP) activities; and were maintained in this state up to 37 passages ( approximately 160 days). Colonies from OVC.EX lines showed classic domed hESC-like morphology surrounded by a ring of fibroblast-like cells, whereas all OVC.ID lines exhibited a mixed cell colony of tightly packed cESCs surrounded by a GATA6+/CDX2- hypoblast-derived support layer. Spontaneous serum-only differentiation without feeder layers demonstrated a strong lineage selection associated with the colony niche type, and not the isolation method. Upon differentiation, cESC lines formed embryoid bodies (EB) comprised of cells representative of all germinal layers, and differentiated into cell types of each layer. Canine ESC lines such as these have the potential to identify differences between embryonic stem cell line derivations, and to develop or to test cell-based transplantation therapies in the dog before attempting human clinical trials.
Collapse
Affiliation(s)
- Jared T Wilcox
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
276
|
Betts DH, Kalionis B. Viable iPSC mice: a step closer to therapeutic applications in humans? Mol Hum Reprod 2009; 16:57-62. [DOI: 10.1093/molehr/gap101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
277
|
Dixon JA, Spinale FG. Large animal models of heart failure: a critical link in the translation of basic science to clinical practice. Circ Heart Fail 2009; 2:262-71. [PMID: 19808348 DOI: 10.1161/circheartfailure.108.814459] [Citation(s) in RCA: 220] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Congestive heart failure (HF) is a clinical syndrome, with hallmarks of fatigue and dyspnea, that continues to be highly prevalent and morbid. Because of the growing burden of HF as the population ages, the need to develop new pharmacological treatments and therapeutic interventions is of paramount importance. Common pathophysiologic features of HF include changes in left ventricle structure, function, and neurohormonal activation. The recapitulation of the HF phenotype in large animal models can allow for the translation of basic science discoveries into clinical therapies. Models of myocardial infarction/ischemia, ischemic cardiomyopathy, ventricular pressure and volume overload, and pacing-induced dilated cardiomyopathy have been created in dogs, pigs, and sheep for the investigation of HF and potential therapies. Large animal models recapitulating the clinical HF phenotype and translating basic science to clinical applications have successfully traveled the journey from bench to bedside. Undoubtedly, large animal models of HF will continue to play a crucial role in the elucidation of biological pathways involved in HF and the development and refinement of HF therapies.
Collapse
Affiliation(s)
- Jennifer A Dixon
- Division of Cardiothoracic Surgery, Medical University of South Carolina and Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC 29425, USA
| | | |
Collapse
|
278
|
Yeung CW, Cheah K, Chan D, Chan BP. Effects of Reconstituted Collagen Matrix on Fates of Mouse Embryonic Stem Cells Before and After Induction for Chondrogenic Differentiation. Tissue Eng Part A 2009; 15:3071-85. [DOI: 10.1089/ten.tea.2008.0661] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Chiu W. Yeung
- Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kathryn Cheah
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Danny Chan
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Barbara P. Chan
- Medical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
279
|
Carpenter MK, Frey-Vasconcells J, Rao MS. Developing safe therapies from human pluripotent stem cells. Nat Biotechnol 2009; 27:606-13. [PMID: 19587662 DOI: 10.1038/nbt0709-606] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
280
|
Gertych A, Wawrowsky KA, Lindsley E, Vishnevsky E, Farkas DL, Tajbakhsh J. Automated quantification of DNA demethylation effects in cells via 3D mapping of nuclear signatures and population homogeneity assessment. Cytometry A 2009; 75:569-83. [PMID: 19459215 DOI: 10.1002/cyto.a.20740] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Today's advanced microscopic imaging applies to the preclinical stages of drug discovery that employ high-throughput and high-content three-dimensional (3D) analysis of cells to more efficiently screen candidate compounds. Drug efficacy can be assessed by measuring response homogeneity to treatment within a cell population. In this study, topologically quantified nuclear patterns of methylated cytosine and global nuclear DNA are utilized as signatures of cellular response to the treatment of cultured cells with the demethylating anti-cancer agents: 5-azacytidine (5-AZA) and octreotide (OCT). Mouse pituitary folliculostellate TtT-GF cells treated with 5-AZA and OCT for 48 hours, and untreated populations, were studied by immunofluorescence with a specific antibody against 5-methylcytosine (MeC), and 4,6-diamidino-2-phenylindole (DAPI) for delineation of methylated sites and global DNA in nuclei (n = 163). Cell images were processed utilizing an automated 3D analysis software that we developed by combining seeded watershed segmentation to extract nuclear shells with measurements of Kullback-Leibler's (K-L) divergence to analyze cell population homogeneity in the relative nuclear distribution patterns of MeC versus DAPI stained sites. Each cell was assigned to one of the four classes: similar, likely similar, unlikely similar, and dissimilar. Evaluation of the different cell groups revealed a significantly higher number of cells with similar or likely similar MeC/DAPI patterns among untreated cells (approximately 100%), 5-AZA-treated cells (90%), and a lower degree of same type of cells (64%) in the OCT-treated population. The latter group contained (28%) of unlikely similar or dissimilar (7%) cells. Our approach was successful in the assessment of cellular behavior relevant to the biological impact of the applied drugs, i.e., the reorganization of MeC/DAPI distribution by demethylation. In a comparison with other metrics, K-L divergence has proven to be a more valuable and robust tool for categorization of individual cells within a population, with potential applications in epigenetic drug screening.
Collapse
Affiliation(s)
- Arkadiusz Gertych
- Minimally Invasive Surgical Technologies Institute, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA.
| | | | | | | | | | | |
Collapse
|
281
|
Pant D, Keefer CL. Expression of Pluripotency-Related Genes during Bovine Inner Cell Mass Explant Culture. CLONING AND STEM CELLS 2009; 11:355-65. [DOI: 10.1089/clo.2008.0078] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Disha Pant
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Carol L. Keefer
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| |
Collapse
|
282
|
Human parthenogenetic embryonic stem cells: one potential resource for cell therapy. ACTA ACUST UNITED AC 2009; 52:599-602. [PMID: 19641863 DOI: 10.1007/s11427-009-0096-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 06/16/2009] [Indexed: 01/10/2023]
Abstract
Pluripotent stem cells derived from somatic cells through such processes as nuclear transfer or induced pluripotent stem (iPS) cells present an important model for biomedical research and provide potential resources for cell replacement therapies. However, the overall efficiency of the conversional nuclear transfer is very low and the safety issue remains a major concern for iPS cells. Embryonic stem cells (ESCs) generated from parthenogenetic embryos are one attractive alternative as a source of histocompatible cells and tissues for cell therapy. Recent studies on human parthenogenetic embryonic stem cells (hPG ESCs) have revealed that these ESCs are very similar to the hESCs derived from IVF or in vivo produced blastocysts in gene expression and other characteristics, but full differentiation and development potential of these hPG ESCs have to be further investigated before clinical research and therapeutic interventions. To generate various pluripotent stem cells, diverse reprogramming techniques and approaches will be developed and integrated. This may help elucidate the fundamental mechanisms underlying reprogramming and stem cell biology, and ultimately benefit cell therapy and regenerative medicine.
Collapse
|
283
|
Fivefold increase in derivation rates of mouse embryonic stem cells after supplementation of the media with multiple factors. Theriogenology 2009; 72:232-42. [DOI: 10.1016/j.theriogenology.2009.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Revised: 02/02/2009] [Accepted: 02/03/2009] [Indexed: 11/18/2022]
|
284
|
Zuk PA. The intracellular distribution of the ES cell totipotent markers OCT4 and Sox2 in adult stem cells differs dramatically according to commercial antibody used. J Cell Biochem 2009; 106:867-77. [PMID: 19199344 DOI: 10.1002/jcb.22054] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
To characterize ES cells, researchers have at their disposal a list of pluripotent markers, such as OCT4. In their quest to determine if adult stem cell populations, such as MSCs and ASCs, are pluripotent, several groups have begun to report the expression of these markers in these cells. Consistent with this, human ASCs (hASCs) are shown in this study to express a plethora of ES pluripotent markers at the gene and protein level, including OCT4, Sox2, and Nanog. When intracellular distribution is examined in hASCs, both OCT4 and Sox2 are expressed within the nuclei of hASCs, consistent with their expression patterns in ES cells. However, a significant amount of expression can be noted within the hASC cytoplasm and a complete absence of nuclear expression is observed for Nanog. Recent descriptions of OCT4 transcript variants may explain the cytoplasmic expression of OCT4 in hASCs and consistent with this, hASCs do express both the OCT4A and 4B transcript variants at the gene level. However, discrepancies arise when these three pluripotent markers are studied at the protein level. Specifically, distinct differences in intracellular expression patterns were noted for OCT4, Sox2, and Nanog from commercial antibody to commercial antibody. These antibody discrepancies persisted when hMSCs and rat ASCs and MSCs were examined. Therefore, confirming the expression of OCT4, Sox2, and Nanog in adult stem cells with today's commercial antibodies must be carefully considered before the designation of pluripotent can be granted.
Collapse
Affiliation(s)
- Patricia A Zuk
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA.
| |
Collapse
|
285
|
WWP2 promotes degradation of transcription factor OCT4 in human embryonic stem cells. Cell Res 2009; 19:561-73. [PMID: 19274063 DOI: 10.1038/cr.2009.31] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
POU transcription factor OCT4 not only plays an essential role in maintaining the pluripotent and self-renewing state of embryonic stem (ES) cells but also acts as a cell fate determinant through a gene dosage effect. However, the molecular mechanisms that control the intracellular OCT4 protein level remain elusive. Here, we report that human WWP2, an E3 ubiquitin (Ub)-protein ligase, interacts with OCT4 specifically through its WW domain and enhances Ub modification of OCT4 both in vitro and in vivo. We first demonstrated that endogenous OCT4 in human ES cells can be post-translationally modified by Ub. Furthermore, we found that WWP2 promoted degradation of OCT4 through the 26S proteasome in a dosage-dependent manner, and the active site cysteine residue of WWP2 was required for both its enzymatic activity and proteolytic effect on OCT4. Remarkably, our data show that the endogenous OCT4 protein level was significantly elevated when WWP2 expression was downregulated by specific RNA interference (RNAi), suggesting that WWP2 is an important regulator for maintaining a proper OCT4 protein level in human ES cells. Moreover, northern blot analysis showed that the WWP2 transcript was widely present in diverse human tissues/organs and highly expressed in undifferentiated human ES cells. However, its expression level was quickly decreased after human ES cells differentiated, indicating that WWP2 expression might be developmentally regulated. Our findings demonstrate that WWP2 is an important regulator of the OCT4 protein level in human ES cells.
Collapse
|
286
|
Beltrami AP, Cesselli D, Beltrami CA. Pluripotency rush! Molecular cues for pluripotency, genetic reprogramming of adult stem cells, and widely multipotent adult cells. Pharmacol Ther 2009; 124:23-30. [PMID: 19545589 DOI: 10.1016/j.pharmthera.2009.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 06/02/2009] [Indexed: 12/14/2022]
Abstract
In the last few years, pluripotent stem cells have been the objective of intense investigation efforts. These cells are of paramount therapeutic interest, since they could be utilized: as in vitro models of disease, for pharmaceutical screening purposes, and for the regeneration of damaged organs. Over the years, pluripotent cells have been cultured from teratomas, the inner cell mass, and primordial germ cells. Accumulating informations have partially decrypted the molecular machinery responsible for the maintenance of a very primitive state, permitting the reprogramming of differentiated cells. Although the debate is still open, an extreme excitement is arising from two strictly related possibilities: pluripotent cells could be obtained from adult tissues with minimal manipulations or very rare pluripotent cells could be identified in adult tissues. This intriguing option will trigger new researches aimed both at identifying the possible biological role of pluripotent adult stem cells and at exploiting their potential clinical use. The present review article will summarize current knowledge of the molecular cues for pluripotency but also discusses whether pluripotent stem cells could be obtained from adult tissues.
Collapse
Affiliation(s)
- Antonio Paolo Beltrami
- Interdepartmental Center for Regenerative Medicine (CIME), Department of Pathology, University of Udine, Italy.
| | | | | |
Collapse
|
287
|
Kim YE, Park JA, Nam KH, Kwon HJ, Lee Y. Pyrrolidine dithiocarbamate-induced activation of ERK and increased expression of c-Fos in mouse embryonic stem cells. BMB Rep 2009; 42:148-53. [PMID: 19336001 DOI: 10.5483/bmbrep.2009.42.3.148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pyrrolidine dithiocarbamate (PDTC) is a stable anti-oxidant or pro-oxidant, depending on the situation, and it is widely used to inhibit the activation of NF-kappaB. We recently reported that PDTC activates the MIP-2 gene in a NF-kappaB-independent and c-Jun-dependent manner in macrophage cells. In this work, we found that PDTC activates signal transduction pathways in mouse ES cells. Among the three different mitogen-activated protein kinase (MAPK) pathways, including the extracellular-signal-regulated kinase (ERK), p38 MAP kinase, and stress-activated protein kinase (SAPK)/Jun N-terminal kinase (JNK) pathways, only the ERK pathway was significantly activated in mouse ES cells after stimulation with PDTC. Additionally, we observed a synergistic activation of ERK and induction of c-Fos after stimulation with PDTC in the presence of mouse embryonic fibroblast (MEF) conditioned medium. In contrast, another NF-kappaB inhibitor, BMS-345541, did not activate the MAP kinase pathways or induce expression of c-Fos. These results suggest that changes in the presence of the NF-kappaB inhibitor PDTC should be carefully considered when it used with mouse ES cells.
Collapse
Affiliation(s)
- Young-Eun Kim
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | | | | | | | | |
Collapse
|
288
|
Induced pluripotent stem cells and the stability of the differentiated state. EMBO Rep 2009; 10:714-21. [PMID: 19543232 DOI: 10.1038/embor.2009.142] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 05/25/2009] [Indexed: 12/16/2022] Open
Abstract
For much of the last century, the differentiated state that characterizes the many cell types of an adult organism was thought to be stable and abrogated only in rare instances by transdifferentiation, metaplasia or cancer. This stability was thought to reside in the autoregulatory molecular circuitry that exists between the cytoplasm and the nucleus, a status quo that could be disrupted during somatic cell nuclear transfer, to reprogramme cells to a pluripotent state. Pioneering work in the 1980s showed that transdifferentiation of cell lineages could be induced by the addition of transcription factors. However, these conversions were usually confined to cell types from the same germ layer, and proof of conversion was difficult to obtain. This deficiency has now been overturned by demonstrations that exogenously added transcription factors can convert differentiated cell types into embryonic-like induced pluripotent stem cells. Here, we highlight the recent progress, and the implications of this work for our understanding of the relationship between the pluripotent and more differentiated cell states.
Collapse
|
289
|
Abstract
For reasons that are unclear the production of embryonic stem cells from ungulates has proved elusive. Here, we describe induced pluripotent stem cells (iPSC) derived from porcine fetal fibroblasts by lentiviral transduction of 4 human (h) genes, hOCT4, hSOX2, hKLF4, and hc-MYC, the combination commonly used to create iPSC in mouse and human. Cells were cultured on irradiated mouse embryonic fibroblasts (MEF) and in medium supplemented with knockout serum replacement and FGF2. Compact colonies of alkaline phosphatase-positive cells emerged after approximately 22 days, providing an overall reprogramming efficiency of approximately 0.1%. The cells expressed porcine OCT4, NANOG, and SOX2 and had high telomerase activity, but also continued to express the 4 human transgenes. Unlike human ESC, the porcine iPSC (piPSC) were positive for SSEA-1, but negative for SSEA-3 and -4. Transcriptional profiling on Affymetrix (porcine) microarrays and real time RT-PCR supported the conclusion that reprogramming to pluripotency was complete. One cell line, ID6, had a normal karyotype, a cell doubling time of approximately 17 h, and has been maintained through >220 doublings. The ID6 line formed embryoid bodies, expressing genes representing all 3 germ layers when cultured under differentiating conditions, and teratomas containing tissues of ectoderm, mesoderm, and endoderm origin in nude mice. We conclude that porcine somatic cells can be reprogrammed to form piPSC. Such cell lines derived from individual animals could provide a means for testing the safety and efficacy of stem cell-derived tissue grafts when returned to the same pigs at a later age.
Collapse
|
290
|
Loh XJ, Gong J, Sakuragi M, Kitajima T, Liu M, Li J, Ito Y. Surface Coating with a Thermoresponsive Copolymer for the Culture and Non-Enzymatic Recovery of Mouse Embryonic Stem Cells. Macromol Biosci 2009; 9:1069-79. [DOI: 10.1002/mabi.200900081] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
291
|
Desbordes SC, Placantonakis DG, Ciro A, Socci ND, Lee G, Djaballah H, Studer L. High-throughput screening assay for the identification of compounds regulating self-renewal and differentiation in human embryonic stem cells. Cell Stem Cell 2009; 2:602-12. [PMID: 18522853 DOI: 10.1016/j.stem.2008.05.010] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 04/01/2008] [Accepted: 05/13/2008] [Indexed: 12/24/2022]
Abstract
High-throughput screening (HTS) of chemical libraries has become a critical tool in basic biology and drug discovery. However, its implementation and the adaptation of high-content assays to human embryonic stem cells (hESCs) have been hampered by multiple technical challenges. Here we present a strategy to adapt hESCs to HTS conditions, resulting in an assay suitable for the discovery of small molecules that drive hESC self-renewal or differentiation. Use of this new assay has led to the identification of several marketed drugs and natural compounds promoting short-term hESC maintenance and compounds directing early lineage choice during differentiation. Global gene expression analysis upon drug treatment defines known and novel pathways correlated to hESC self-renewal and differentiation. Our results demonstrate feasibility of hESC-based HTS and enhance the repertoire of chemical compounds for manipulating hESC fate. The availability of high-content assays should accelerate progress in basic and translational hESC biology.
Collapse
Affiliation(s)
- Sabrina C Desbordes
- Developmental Biology Program, Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | | | | | | | | | | | |
Collapse
|
292
|
Momose Y, Matsunaga T, Murai K, Takezawa T, Ohmori S. Differentiation of monkey embryonic stem cells into hepatocytes and mRNA expression of cytochrome p450 enzymes responsible for drug metabolism: comparison of embryoid body formation conditions and matrices. Biol Pharm Bull 2009; 32:619-26. [PMID: 19336894 DOI: 10.1248/bpb.32.619] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated the effects of embryoid body (EB) forming conditions on the expression of hepatocyte marker genes such as alpha-fetoprotein, albumin and CYP7A1 in cells cultured on Matrigel-coated plates for 15 d. The expression levels of hepatocyte marker genes in the cells cultured for 2 d for EB formation from cynomolgus monkey embryonic stem (cmES) cells was higher than those in cells cultured for 5 d. However, the fragment-size of cmES colonies did not markedly affect the expression levels. The expression levels of hepatocyte marker genes, and CYP1A1 and CYP2C43 in cells cultured on Matrigel were considerably higher than those on Matrigel reduced and collagen I. CYP1A1 and CYP3A8 mRNAs were significantly induced by 3-methylcholanthrene and rifampicin, respectively. However, CYP2C43 and CYP2D17 were not induced by these compounds. These results suggested that the differentiation into hepatocytes is affected by the incubation period for EB formation, and that Matrigel successfully promoted in vitro differentiation of cmES cells to hepatocytes.
Collapse
|
293
|
Krtolica A, Ilic D, Genbacev O, Miller RK. Human embryonic stem cells as a model for embryotoxicity screening. Regen Med 2009; 4:449-59. [DOI: 10.2217/rme.09.13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Reproductive toxicity encompasses harmful effects of various agents on all aspects and stages of the reproductive cycle, including infertility and the induction of adverse effects in the embryo/fetus. In developing a model for reproductive toxicity screening, it is important to define the stage of the human reproductive cycle that this specific model is going to recreate in vitro and to identify molecular targets that are critical for this stage of development. In this review, we focus our discussion on modeling pre-implantation embryotoxicity. The rationale for this is that despite advances on both clinical and biological levels, many unresolved infertility cases may be due to our lack of knowledge regarding environmental influences on this short, but critical stage of development. Data from in vitro fertilization practice suggest that the early-dividing embryo is very sensitive to numerous factors present in its microenvironment. In vivo, as the embryo travels down the oviduct, physical or chemical insults can directly damage the embryo and/or prevent implantation, and cause infertility. Multiple lines of evidence point to the differences between mouse and human pre-implantation development and between mouse and human embryonic stem cells (hESCs). In light of these data we present the case that hESCs and their derivatives are better suited as in vitro models for human pre-implantation development than their mouse counterparts. We then describe some of the most promising hESC-based systems that are used today to model certain aspects of development in the human pre-implantation embryo and that have the potential to be used for embryo toxicity screening tests in the near future. Described systems model two major events during differentiation of the human pre-implantation embryo: differentiation of the trophectoderm and segregation of the inner cell mass into epiblast and hypoblast. The first event is replicated in vitro by triggering either direct or indirect (through embryoid body stage) differentiation into trophectoderm. The second event can be modeled using the recently described system of high-throughput generation of embryoid bodies that recapitulate segregation of inner cell mass. We conclude by discussing the potential of these existing models in toxicology studies and the possibilities for their improvement in the future.
Collapse
Affiliation(s)
- Ana Krtolica
- SLL Sciences, StemLifeLine, Inc., San Carlos, CA, USA
| | - Dusko Ilic
- SLL Sciences, StemLifeLine, Inc., San Carlos, CA, USA
| | - Olga Genbacev
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Francisco, 513 Parnassus Ave., San Francisco, CA, USA
| | - Richard K Miller
- School of Medicine & Dentistry, University of Rochester, NY, USA
| |
Collapse
|
294
|
Bratt-Leal AM, Carpenedo RL, McDevitt TC. Engineering the embryoid body microenvironment to direct embryonic stem cell differentiation. Biotechnol Prog 2009; 25:43-51. [PMID: 19198003 DOI: 10.1002/btpr.139] [Citation(s) in RCA: 210] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Embryonic stem cells (ESCs) are pluripotent cells capable of differentiating into all somatic and germ cell types. The intrinsic ability of pluripotent cells to generate a vast array of different cells makes ESCs a robust resource for a variety of cell transplantation and tissue engineering applications, however, efficient and controlled means of directing ESC differentiation is essential for the development of regenerative therapies. ESCs are commonly differentiated in vitro by spontaneously self-assembling in suspension culture into 3D cell aggregates called embryoid bodies (EBs), which mimic many of the hallmarks of early embryonic development, yet the 3D organization and structure of EBs also presents unique challenges to effectively direct the differentiation of the cells. ESC differentiation is strongly influenced by physical and chemical signals comprising the local extracellular microenvironment, thus current methods to engineer EB differentiation have focused primarily on spatially controlling EB size, adding soluble factors to the media, or culturing EBs on or within natural or synthetic extracellular matrices. Although most such strategies aim to influence differentiation from the exterior of EBs, engineering the microenvironment directly within EBs enables new opportunities to efficiently direct the fate of the cells by locally controlling the presentation of morphogenic cues.
Collapse
Affiliation(s)
- Andrés M Bratt-Leal
- The Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | | | | |
Collapse
|
295
|
Zhao X, Ruan Y, Wei CL. Tackling the epigenome in the pluripotent stem cells. J Genet Genomics 2009; 35:403-12. [PMID: 18640620 DOI: 10.1016/s1673-8527(08)60058-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 05/23/2008] [Accepted: 05/24/2008] [Indexed: 11/26/2022]
Abstract
Embryonic stem cells are unique in their abilities of self-renewal and to differentiate into many, if not all, cellular lineages. Transcriptional regulation, epigenetic modifications and chromatin structures are the key modulators in controlling such pluripotency nature of embryonic stem cell genomes, particularly in the developmental decisions and the maintenance of cell fates. Among them, epigenetic regulation of gene expression is mediated partly by covalent modifications of core histone proteins including methylation, phosphorylation and acetylation. Moreover, the chromatins in stem cell genome appear as a highly organized structure containing distinct functional domains. Recent rapid progress of new technologies enables us to take a global, unbiased and comprehensive view of the epigenetic modifications and chromatin structures that contribute to gene expression regulation and cell identity during diverse developmental stages. Here, we summarized the latest advances made by high throughput approaches in profiling epigenetic modifications and chromatin conformations, with an emphasis on genome-wide analysis of histone modifications and their implications in pluripotency nature of embryonic stem cells.
Collapse
Affiliation(s)
- Xiaodong Zhao
- Genome Technology and Biology Group, Genome Institute of Singapore, 138672, Singapore
| | | | | |
Collapse
|
296
|
Asynchronous replication and autosome-pair non-equivalence in human embryonic stem cells. PLoS One 2009; 4:e4970. [PMID: 19325893 PMCID: PMC2657208 DOI: 10.1371/journal.pone.0004970] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 02/26/2009] [Indexed: 12/02/2022] Open
Abstract
A number of mammalian genes exhibit the unusual properties of random monoallelic expression and random asynchronous replication. Such exceptional genes include genes subject to X inactivation and autosomal genes including odorant receptors, immunoglobulins, interleukins, pheromone receptors, and p120 catenin. In differentiated cells, random asynchronous replication of interspersed autosomal genes is coordinated at the whole chromosome level, indicative of chromosome-pair non-equivalence. Here we have investigated the replication pattern of the random asynchronously replicating genes in undifferentiated human embryonic stem cells, using fluorescence in situ hybridization based assay. We show that allele-specific replication of X-linked genes and random monoallelic autosomal genes occur in human embryonic stem cells. The direction of replication is coordinated at the whole chromosome level and can cross the centromere, indicating the existence of autosome-pair non-equivalence in human embryonic stem cells. These results suggest that epigenetic mechanism(s) that randomly distinguish between two parental alleles are emerging in the cells of the inner cell mass, the source of human embryonic stem cells.
Collapse
|
297
|
Lavial F, Acloque H, Bachelard E, Nieto MA, Samarut J, Pain B. Ectopic expression of Cvh (Chicken Vasa homologue) mediates the reprogramming of chicken embryonic stem cells to a germ cell fate. Dev Biol 2009; 330:73-82. [PMID: 19324033 DOI: 10.1016/j.ydbio.2009.03.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 02/19/2009] [Accepted: 03/13/2009] [Indexed: 10/21/2022]
Abstract
When they are derived from blastodermal cells of the pre-primitive streak in vitro, the pluripotency of Chicken Embryonic Stem Cells (cESC) can be controlled by the cPouV and Nanog genes. These cESC can differentiate into derivatives of the three germ layers both in vitro and in vivo, but they only weakly colonize the gonads of host embryos. By contrast, non-cultured blastodermal cells and long-term cultured chicken primordial germ cells maintain full germline competence. This restriction in the germline potential of the cESC may result from either early germline determination in the donor embryos or it may occur as a result of in vitro culture. We are interested in understanding the genetic determinants of germline programming. The RNA binding protein Cvh (Chicken Vasa Homologue) is considered as one such determinant, although its role in germ cell physiology is still unclear. Here we show that the exogenous expression of Cvh, combined with appropriate culture conditions, induces cESC reprogramming towards a germ cell fate. Indeed, these cells express the Dazl, Tudor and Sycp3 germline markers, and they display improved germline colonization and adopt a germ cell fate when injected into recipient embryos. Thus, our results demonstrate that Vasa can drive ES cell differentiation towards the germ cell lineage, both in vitro and in vivo.
Collapse
Affiliation(s)
- Fabrice Lavial
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, UMR 5242, INRA, Ecole Normale Supérieure de Lyon, France
| | | | | | | | | | | |
Collapse
|
298
|
Farin A, Liu CY, Elder JB, Langmoen IA, Apuzzo MLJ. The biological restoration of central nervous system architecture and function: part 1-foundations and historical landmarks in contemporary stem cell biology. Neurosurgery 2009; 64:15-39; discussion 34. [PMID: 19145154 DOI: 10.1227/01.neu.0000337580.02706.dc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Since their discovery, stem cells have fascinated scientists with their ultimate potential: the ability to cure disease, repair altered physiology, and reverse neurological deficit. Stem cell science unquestionably promises to eliminate many of the tragic limitations contemporary medicine must acknowledge, and cloning may provide young cells for an aging population. Although it is widely believed that stem cells will transform the way medicine is practiced, therapeutic interventions using stem cell technology are still in their infancy. The 3 most common stem cell sources studied today are umbilical cord blood, bone marrow, and human embryos. Although cord blood is currently used to treat dozens of disorders and bone marrow stem cells have been used clinically since the 1960s, human embryonic stem cells have yet to be successfully applied to any disease. Undeniably, stem cell therapy has the potential to be one of the most powerful therapeutic options available. In this introductory article of a 5-part series on stem cells, we narrate the evolution of modern stem cell science, delineating major landmarks that will prove responsible for taking stem cell technology from the laboratory into revolutionary clinical applications: from the first milestone of identifying the mouse hematopoietic stem cell to the latest feats of producing pluripotent stem cells without embryos at all. In Part 2, we present the evidence demonstrating the certainty of adult mammalian neurogenesis; in Parts 3 and 4, we describe neurosurgical applications of stem cell technology; and in Part 5, we discuss the philosophical and ethical issues surrounding stem cell therapy, as well as future areas of exploration.
Collapse
Affiliation(s)
- Azadeh Farin
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | |
Collapse
|
299
|
Wu D, Pang Y, Ke Y, Yu J, He Z, Tautz L, Mustelin T, Ding S, Huang Z, Feng GS. A conserved mechanism for control of human and mouse embryonic stem cell pluripotency and differentiation by shp2 tyrosine phosphatase. PLoS One 2009; 4:e4914. [PMID: 19290061 PMCID: PMC2655646 DOI: 10.1371/journal.pone.0004914] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 02/10/2009] [Indexed: 12/31/2022] Open
Abstract
Recent studies have suggested distinctive biological properties and signaling mechanisms between human and mouse embryonic stem cells (hESCs and mESCs). Herein we report that Shp2, a protein tyrosine phosphatase with two SH2 domains, has a conserved role in orchestration of intracellular signaling cascades resulting in initiation of differentiation in both hESCs and mESCs. Homozygous deletion of Shp2 in mESCs inhibited differentiation into all three germ layers, and siRNA-mediated knockdown of Shp2 expression in hESCs led to a similar phenotype of impaired differentiation. A small molecule inhibitor of Shp2 enzyme suppressed both hESC and mESC differentiation capacity. Shp2 modulates Erk, Stat3 and Smad pathways in ES cells and, in particular, Shp2 regulates BMP4-Smad pathway bi-directionally in mESCs and hESCs. These results reveal a common signaling mechanism shared by human and mouse ESCs via Shp2 modulation of overlapping and divergent pathways.
Collapse
Affiliation(s)
- Dongmei Wu
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Yuhong Pang
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Yuehai Ke
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Jianxiu Yu
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Zhao He
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Lutz Tautz
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Tomas Mustelin
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Sheng Ding
- Department of Chemistry, Scripps Research Institute, La Jolla, California, United States of American
| | - Ziwei Huang
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
| | - Gen-Sheng Feng
- Programs in Signal Transduction, and Stem Cells and Regeneration, Burnham Institute for Medical Research, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
300
|
Müller T, Fleischmann G, Eildermann K, Mätz-Rensing K, Horn PA, Sasaki E, Behr R. A novel embryonic stem cell line derived from the common marmoset monkey (Callithrix jacchus) exhibiting germ cell-like characteristics. Hum Reprod 2009; 24:1359-72. [PMID: 19251728 DOI: 10.1093/humrep/dep012] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Embryonic stem cells (ESC) hold great promise for the treatment of degenerative diseases. However, before clinical application of ESC in cell replacement therapy can be achieved, the safety and feasibility must be extensively tested in animal models. The common marmoset monkey (Callithrix jacchus) is a useful preclinical non-human primate model due to its physiological similarities to human. Yet, few marmoset ESC lines exist and differences in their developmental potential remain unclear. METHODS Blastocysts were collected and immunosurgery was performed. cjes001 cells were tested for euploidy by karyotyping. The presence of markers for pluripotency was confirmed by immunofluorescence staining and RT-PCR. Histology of teratoma, in vitro differentiation and embryoid body formation revealed the differentiation potential. RESULTS cjes001 cells displayed a normal 46,XX karyotype. Alkaline phosphatase activity, expression of telomerase and the transcription factors OCT4, NANOG and SOX2 as well as the presence of stage-specific embryonic antigen (SSEA)-3, SSEA-4, tumor rejection antigens (TRA)-1-60, and TRA-1-81 indicated pluripotency. Teratoma formation assay displayed derivatives of all three embryonic germ layers. Upon non-directed differentiation, the cells expressed the germ cell markers VASA, BOULE, germ cell nuclear factor and synaptonemal complex protein 3 and showed co-localization of VASA protein within individual cells with the germ line stem cell markers CD9, CD49f, SSEA-4 and protein gene product 9.5, respectively. CONCLUSIONS The cjes001 cells represent a new pluripotent ESC line with evidence for enhanced spontaneous differentiation potential into germ cells. This cjes001 line will be very valuable for comparative studies on primate ESC biology.
Collapse
Affiliation(s)
- Thomas Müller
- Stem Cell Research Group, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|