Journal articles on the topic 'Embryonic stem cells Differentiation'

To see the other types of publications on this topic, follow the link: Embryonic stem cells Differentiation.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Embryonic stem cells Differentiation.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Challa, Stalin Reddy, and Swathi Goli. "Differentiation of Human Embryonic Stem Cells into Engrafting Myogenic Precursor Cells." Stem cell Research and Therapeutics International 1, no. 1 (April 16, 2019): 01–05. http://dx.doi.org/10.31579/2643-1912/002.

Full text
Abstract:
Degenerative muscle diseases affect muscle tissue integrity and function. Human embryonic stem cells (hESC) are an attractive source of cells to use in regenerative therapies due to their unlimited capacity to divide and ability to specialize into a wide variety of cell types. A practical way to derive therapeutic myogenic stem cells from hESC is lacking. In this study, we demonstrate the development of two serum-free conditions to direct the differentiation of hESC towards a myogenic precursor state. Using TGFß and PI3Kinase inhibitors in combination with bFGF we showed that one week of differentiation is sufficient for hESC to specialize into PAX3+/PAX7+ myogenic precursor cells. These cells also possess the capacity to further differentiate in vitro into more specialized myogenic cells that express MYOD, Myogenin, Desmin and MYHC, and showed engraftment in vivo upon transplantation in immunodeficient mice. Ex vivo myomechanical studies of dystrophic mouse hindlimb muscle showed functional improvement one month post-transplantation. In summary, this study describes a promising system to derive engrafting muscle precursor cells solely using chemical substances in serum-free conditions and without genetic manipulation.
APA, Harvard, Vancouver, ISO, and other styles
2

Okabe, Shigeo. "Differentiation of Embryonic Stem Cells." Current Protocols in Neuroscience 00, no. 1 (September 1997): 3.6.1–3.6.13. http://dx.doi.org/10.1002/0471142301.ns0306s00.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Pera, M. F., B. Reubinoff, and A. Trounson. "Human embryonic stem cells." Journal of Cell Science 113, no. 1 (January 1, 2000): 5–10. http://dx.doi.org/10.1242/jcs.113.1.5.

Full text
Abstract:
Embryonic stem (ES) cells are cells derived from the early embryo that can be propagated indefinitely in the primitive undifferentiated state while remaining pluripotent; they share these properties with embryonic germ (EG) cells. Candidate ES and EG cell lines from the human blastocyst and embryonic gonad can differentiate into multiple types of somatic cell. The phenotype of the blastocyst-derived cell lines is very similar to that of monkey ES cells and pluripotent human embryonal carcinoma cells, but differs from that of mouse ES cells or the human germ-cell-derived stem cells. Although our understanding of the control of growth and differentiation of human ES cells is quite limited, it is clear that the development of these cell lines will have a widespread impact on biomedical research.
APA, Harvard, Vancouver, ISO, and other styles
4

Dani, C., A. G. Smith, S. Dessolin, P. Leroy, L. Staccini, P. Villageois, C. Darimont, and G. Ailhaud. "Differentiation of embryonic stem cells into adipocytes in vitro." Journal of Cell Science 110, no. 11 (June 1, 1997): 1279–85. http://dx.doi.org/10.1242/jcs.110.11.1279.

Full text
Abstract:
Embryonic stem cells, derived from the inner cell mass of murine blastocysts, can be maintained in a totipotent state in vitro. In appropriate conditions embryonic stem cells have been shown to differentiate in vitro into various derivatives of all three primary germ layers. We describe in this paper conditions to induce differentiation of embryonic stem cells reliably and at high efficiency into adipocytes. A prerequisite is to treat early developing embryonic stem cell-derived embryoid bodies with retinoic acid for a precise period of time. Retinoic acid could not be substituted by adipogenic hormones nor by potent activators of peroxisome proliferator-activated receptors. Treatment with retinoic acid resulted in the subsequent appearance of large clusters of mature adipocytes in embryoid body outgrowths. Lipogenic and lipolytic activities as well as high level expression of adipocyte specific genes could be detected in these cultures. Analysis of expression of potential adipogenic genes, such as peroxisome proliferator-activated receptors gamma and delta and CCAAT/enhancer binding protein beta, during differentiation of retinoic acid-treated embryoid bodies has been performed. The temporal pattern of expression of genes encoding these nuclear factors resembled that found during mouse embryogenesis. The differentiation of embryonic stem cells into adipocytes will provide an invaluable model for the characterisation of the role of genes expressed during the adipocyte development programme and for the identification of new adipogenic regulatory genes.
APA, Harvard, Vancouver, ISO, and other styles
5

ORLOVSKAYA, I., I. SCHRAUFSTATTER, J. LORING, and S. KHALDOYANIDI. "Hematopoietic differentiation of embryonic stem cells." Methods 45, no. 2 (June 2008): 159–67. http://dx.doi.org/10.1016/j.ymeth.2008.03.002.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Bradley, A. "Embryonic stem cells: Proliferation and differentiation." Current Opinion in Cell Biology 2, no. 6 (December 1990): 1013–17. http://dx.doi.org/10.1016/0955-0674(90)90150-d.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

van Inzen, Wouter G., Maikel P. Peppelenbosch, Maria W. M. van den Brand, Leon G. J. Tertoolen, and Siegfried W. de Laat. "Neuronal differentiation of embryonic stem cells." Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 1312, no. 1 (June 1996): 21–26. http://dx.doi.org/10.1016/0167-4889(96)00011-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Shinde, Vaibhav, Sonja Brungs, Margit Henry, Lucia Wegener, Harshal Nemade, Tamara Rotshteyn, Aviseka Acharya, et al. "Simulated Microgravity Modulates Differentiation Processes of Embryonic Stem Cells." Cellular Physiology and Biochemistry 38, no. 4 (2016): 1483–99. http://dx.doi.org/10.1159/000443090.

Full text
Abstract:
Background/Aims: Embryonic developmental studies under microgravity conditions in space are very limited. To study the effects of altered gravity on the embryonic development processes we established an in vitro methodology allowing differentiation of mouse embryonic stem cells (mESCs) under simulated microgravity within a fast-rotating clinostat (clinorotation) and capture of microarray-based gene signatures. Methods: The differentiating mESCs were cultured in a 2D pipette clinostat. The microarray and bioinformatics tools were used to capture genes that are deregulated by simulated microgravity and their impact on developmental biological processes. Results: The data analysis demonstrated that differentiation of mESCs in pipettes for 3 days resultet to early germ layer differentiation and then to the different somatic cell types after further 7 days of differentiation in the Petri dishes. Clinorotation influences differentiation as well as non-differentiation related biological processes like cytoskeleton related 19 genes were modulated. Notably, simulated microgravity deregulated genes Cyr61, Thbs1, Parva, Dhrs3, Jun, Tpm1, Fzd2 and Dll1 are involved in heart morphogenesis as an acute response on day 3. If the stem cells were further cultivated under normal gravity conditions (1 g) after clinorotation, the expression of cardiomyocytes specific genes such as Tnnt2, Rbp4, Tnni1, Csrp3, Nppb and Mybpc3 on day 10 was inhibited. This correlated well with a decreasing beating activity of the 10-days old embryoid bodies (EBs). Finally, we captured Gadd45g, Jun, Thbs1, Cyr61and Dll1 genes whose expressions were modulated by simulated microgravity and by real microgravity in various reported studies. Simulated microgravity also deregulated genes belonging to the MAP kinase and focal dhesion signal transduction pathways. Conclusion: One of the most prominent biological processes affected by simulated microgravity was the process of cardiomyogenesis. The most significant simulated microgravity-affected genes, signal transduction pathways, and biological processes which are relevant for mESCs differentiation have been identified and discussed below.
APA, Harvard, Vancouver, ISO, and other styles
9

Kowalski, Michael P., Amy Yoder, Li Liu, and Laura Pajak. "Controlling Embryonic Stem Cell Growth and Differentiation by Automation." Journal of Biomolecular Screening 17, no. 9 (August 15, 2012): 1171–79. http://dx.doi.org/10.1177/1087057112452783.

Full text
Abstract:
Despite significant use in basic research, embryonic stem cells have just begun to be used in the drug discovery process. Barriers to the adoption of embryonic stem cells in drug discovery include the difficulty in growing cells and inconsistent differentiation to the desired cellular phenotype. Embryonic stem cell cultures require consistent and frequent handling to maintain the cells in a pluripotent state. In addition, the preferred hanging drop method of embryoid body (EB) differentiation is not amenable to high-throughput methods, and suspension cultures of EBs show a high degree of variability. Murine embryonic stem cells passaged on an automated platform maintained ≥90% viability and pluripotency. We also developed a method of EB formation using 384-well microplates that form a single EB per well, with excellent uniformity across EBs. This format facilitated high-throughput differentiation and enabled screens to optimize directed differentiation into a desired cell type. Using this approach, we identified conditions that enhanced cardiomyocyte differentiation sevenfold. This optimized differentiation method showed excellent consistency for such a complex biological process. This automated approach to embryonic stem cell handling and differentiation can provide the high and consistent yields of differentiated cell types required for basic research, compound screens, and toxicity studies.
APA, Harvard, Vancouver, ISO, and other styles
10

Li, Xiuju, Pratap Karki, Lei Lei, Huayan Wang, and Larry Fliegel. "Na+/H+ exchanger isoform 1 facilitates cardiomyocyte embryonic stem cell differentiation." American Journal of Physiology-Heart and Circulatory Physiology 296, no. 1 (January 2009): H159—H170. http://dx.doi.org/10.1152/ajpheart.00375.2008.

Full text
Abstract:
Embryonic stem cells provide one potential source of cardiomyocytes for cardiac transplantation; however, after differentiation of stem cells in vitro, cardiomyocytes usually account for only a minority of cells present. To gain insights into improving cardiomyocyte development from stem cells, we examined the role of the Na+/H+ exchanger isoform 1 (NHE1) in cardiomyocyte differentiation. NHE1 protein and message levels were induced by treatment of CGR8 cells to form embryoid bodies and cardiomyocytes. The NHE1 protein was present on the cell surface and NHE1 inhibitor-sensitive activity was detected. Inhibition of NHE1 activity during differentiation of CGR8 cells prevented cardiomyocyte differentiation as indicated by decreased message for transcription factors Nkx2-5 and Tbx5 and decreased levels of α-myosin heavy chain protein. Increased expression of NHE1 from an adenoviral vector facilitated cardiomyocyte differentiation. Similar results were found with cardiomyocyte differentiation of P19 embryonal carcinoma cells. CGR8 cells were treated to induce differentiation, but when differentiation was inhibited by dispersing the EBs, myocardial development was inhibited. The results demonstrate that NHE1 activity is important in facilitating stem cell differentiation to cardiomyocyte lineage. Elevated NHE1 expression appears to be triggered as part of the process that facilitates cardiomyocyte development.
APA, Harvard, Vancouver, ISO, and other styles
11

Olivier, Emmanuel N., Anne C. Rybicki, and Eric E. Bouhassira. "Differentiation of Human Embryonic Stem Cells into Mesenchymal Stem Cells." Blood 106, no. 11 (November 16, 2005): 1389. http://dx.doi.org/10.1182/blood.v106.11.1389.1389.

Full text
Abstract:
Abstract Mesenchymal stem cells (MSC) are multipotent progenitors that contribute to the formation of many connective tissues including fat, bone, cartilage and muscle. Because of this great versatility MSCs have a large therapeutic potential particularly in the areas of cell therapy and regenerative and reconstructive medicine. MSCs are rare cells that can be isolated from tissues such as bone marrow, cartilage and muscles. Since no unique marker characteristic of MSCs has been identified, investigators have relied on a series of functional and morphological criteria to identify them. These criteria include growth on plastic, resistance to trypsin, presence of specific cell surface antigens and potential to differentiate into adipocytes, chondrocytes and osteoblasts.We report here a method to reproducibly differentiate human embryonic stem cells (hESCs) into MSCs by pre-differentiating for 8 days hESCs growing on MEF, mechanically dissociating the differentiated colonies, replating the differentiated colonies in DMEM, 10% FBS and 7.5% CO2 for 4 to 8 weeks until a thick multi-layer epithelium-like sheet of cells develop; and dissociating these multi-layer structures with a combination of proteases. The cells obtained with this procedure are morphologically similar to MSCs, are contact-inhibited, can be grown in culture for about 20–25 passages and have a gene expression profile, as determined by cDNA micro-array, similar to published profiles of mesenchymal stem cells. Immuno-phenotyping of these hESC-derived MSCs revealed a immuno-profile similar to bone marrow MSCs (negative for CD34, CD45 and positive for CD13, CD44, CD71, CD73, CD105, CD166, HLA ABC, SSEA4 and TRA1-85). Functional differentiation experiments revealed that hESC-derived MSCs can be differentiated into osteocytes and adipocytes as demonstrated by staining with alizarin red and oil red O. Finally, we have also shown that hESC-derived MSCs can support the growth of undifferentiated hESCs cells and of CD34+ hematopoietic cells.The ability to produce MSCs from hESCs should prove useful to produce large amount of genetically identical and genetically modifiable MSCS that can be used to study the biology of MSCs and for therapeutic applications.
APA, Harvard, Vancouver, ISO, and other styles
12

Kanaji, Taisuke, Takashi Okamura, and Peter J. Newman. "Filamin A Controls Embryonic Stem Cell Differentiation." Blood 114, no. 22 (November 20, 2009): 4599. http://dx.doi.org/10.1182/blood.v114.22.4599.4599.

Full text
Abstract:
Abstract Abstract 4599 Filamin A is a major non-muscle actin binding protein that plays an important role in cross-linking cortical actin filaments into three-dimensional networks. In addition to its role as a cytoskeletal scaffolding molecule, Filamin A is also known to bind more than 30 other proteins, regulating their subcellular location and coordinating their ability to signal. To analyze the role of filamin A in mouse embryonic stem (ES) cell maturation, we generated filamin ALow ES cells by introducing a micro-RNA that specifically downregulates filamin A expression under the control of a cytomegalovirus promoter. Filamin ALow ES cells exhibited a more rounded morphology than did their wild-type filamin ANormal counterparts, and expressed increased levels of the ES cell transcription factor Nanog. In contrast, non-transfected cells in the same culture dish retained normal expression of filamin A, expressed low levels of Nanog, and exhibited a more elongated and spread phenotype characteristic of differentiating cells. Further evidence for a role for filamin A in ES cell differentiation was provided by the observation that withdrawing leukemia inhibitory factor (LIF) to induce ES cell differentiation was accompanied by increased expression of filamin A, a concomitant loss of Nanog expression, and acquisition of a differentiated morphology. Filamin ALow ES cells were able to retain their undifferentiated phenotype, as evaluated by alkaline phosphatase (Alp) activity, in the presence of a 10-fold lower concentration of LIF than was permissive for filamin ANormal ES cells, or following exposure to the differentiating agent, bone morphogenic protein 4 (BMP4). LIF-induced phosphorylation of ERK was decreased in filamin ALow relative to filamin ANormal ES cells, as was BMP-induced phosphorylation of Smad1/5 - two signaling pathways that initiate ES cell differentiation. Finally, embryoid bodies comprised of filamin ALow ES cells were unable to differentiate into CD41+ hematopoietic progenitor cells. Taken together, these data demonstrate that filamin A plays a previously unrecognized, but critical, scaffolding function that support both the LIF - ERK and BMP4 - Smad1/5 signaling pathways leading to ES and hematopoietic cell differentiation. Manipulation of filamin levels might be useful in the future to modulate the differentiation requirements for a variety of clinically-and therapeutically-useful stem cells. Disclosures: Newman: Novo Nordisk: Consultancy; New York Blood Center: Membership on an entity's Board of Directors or advisory committees.
APA, Harvard, Vancouver, ISO, and other styles
13

Wakao, Hiroshi. "Differentiation of iNKT Cells from Embryonic Stem Cells." Current Immunology Reviews 6, no. 2 (May 1, 2010): 102–8. http://dx.doi.org/10.2174/157339510791111754.

Full text
APA, Harvard, Vancouver, ISO, and other styles
14

Tao, Hongyan, Xiaoniao Chen, Anbang Wei, Xianghe Song, Weiqiang Wang, Lu Liang, Qinjun Zhao, et al. "Comparison of Teratoma Formation between Embryonic Stem Cells and Parthenogenetic Embryonic Stem Cells by Molecular Imaging." Stem Cells International 2018 (March 25, 2018): 1–9. http://dx.doi.org/10.1155/2018/7906531.

Full text
Abstract:
With their properties of self-renewal and differentiation, embryonic stem (ES) cells hold great promises for regenerative therapy. However, teratoma formation and ethical concerns of ES cells may restrict their potential clinical applications. Currently, parthenogenetic embryonic stem (pES) cells have attracted the interest of researchers for its self-renewing and pluripotent differentiation while eliciting less ethic concerns. In this study, we established a model with ES and pES cells both stably transfected with a double-fusion reporter gene containing renilla luciferase (Rluc) and red fluorescent protein (RFP) to analyze the mechanisms of teratoma formation. Transgenic Vegfr2-luc mouse, which expresses firefly luciferase (Fluc) under the promoter of vascular endothelial growth factor receptor 2 (Vegfr2-luc), was used to trace the growth of new blood vessel recruited by transplanted cells. Bioluminescence imaging (BLI) of Rluc/Fluc provides an effective tool in estimating the growth and angiogenesis of teratoma in vivo. We found that the tumorigenesis and angiogenesis capacity of ES cells were higher than those of pES cells, in which VEGF/VEGFR2 signal pathway plays an important role. In conclusion, pES cells have the decreased potential of teratoma formation but meanwhile have similar differentiating capacity compared with ES cells. These data demonstrate that pES cells provide an alternative source for ES cells with the risk reduction of teratoma formation and without ethical controversy.
APA, Harvard, Vancouver, ISO, and other styles
15

Zhu, Jian, Yitian Wang, Wei Yu, Kaishun Xia, Yuluan Huang, Junjie Wang, Bing Liu, Huimin Tao, Chengzhen Liang, and Fangcai Li. "Long Noncoding RNA: Function and Mechanism on Differentiation of Mesenchymal Stem Cells and Embryonic Stem Cells." Current Stem Cell Research & Therapy 14, no. 3 (April 29, 2019): 259–67. http://dx.doi.org/10.2174/1574888x14666181127145809.

Full text
Abstract:
Background:Long suspected as transcriptional noise, recently recognized, long non-coding RNAs (lncRNAs) are emerging as an indicator, biomarker and therapy target in the physiologic and pathologic process. Mesenchymal stem cells and embryonic stem cells are important source for normal and therapeutic tissue repair. However, the mechanism of stem cell differentiation is not completely understood. Research on lncRNAs may provide novel insights into the mechanism of differentiation process of the stem cell which is important for the application of stem cell therapy. The lncRNAs field is still very young, new insights into lncRNAs function are emerging to a greater understanding of biological processes. Objective: In this review, we summarize the recent researches studying lncRNAs and illustrate how they act in the differentiation of the mesenchymal stem cells and embryonic stem cells, and discuss some future directions in this field. Results: Numerous lncRNAs were differentially expressed during differentiation of mesenchymal stem cells and embryonic stem cells. LncRNAs were able to regulate the differentiation processes through epigenetic regulation, transcription regulation and post-transcription regulation. Conclusion: LncRNAs are involved in the differentiation process of mesenchymal stem cells and embryonic stem cells, and they could become promising indicator, biomarker and therapeutic targets in the physiologic and pathologic process. However, the mechanisms of the role of lncRNAs still require further investigation.
APA, Harvard, Vancouver, ISO, and other styles
16

Hong, S., J. K. Kang, C. J. Bae, E. S. Ryu, S. H. Lee, and J. H. Lee. "Development of Efficient Cardiac Differentiation Method of Mouse Embryonic Stem Cells." Key Engineering Materials 342-343 (July 2007): 25–28. http://dx.doi.org/10.4028/www.scientific.net/kem.342-343.25.

Full text
Abstract:
To obtain an enhanced population of cardiomyocytes from differentiating mouse embryonic stem (ES) cells, we confirmed the role of noggin treatment during the cardiac differentiation of mouse ES cells. ES cells were cultured in ES medium containing both noggin and LIF for 3 days on the mouse embryonic fibroblast feeder layer, followed by dissociated and suspension culture without LIF to form the embryoid body (EB). The next day, noggin was eliminated and EBs were cultured continuously. Noggin treated ES cells showed a relatively rapid increase of cardiac marker genes, while the vehicle (PBS) treated group showed no significant cardiac marker expression at 4 days after the EB formation. Furthermore, Noggin treated ES cells showed 68.00±9.16% spontaneous beating EBs at 12 days after the EB formation. To develop a more efficient cardiomyocyte differentiation method, we tested several known cardiogenic reagents (ascorbic acid, 5’-Azacytidine, LiCl, oxytocin, FGF2 and PDGF-BB) after noggin induction or we cultured noggin treated ES cells on various extracellular matrixes (collagen, fibronectin and Matrigel). Quantitative RT-PCR and immunocytochemistry results showed a significantly increased cardiac differentiation rate in the FGF2 treated group. Differentiation on the collagen extracellular matrix (ECM) could slightly increase the cardiac differentiation efficiency. These results show the possibilities for the establishment of selective differentiation conditions for the cardiac differentiation of mouse ES cells.
APA, Harvard, Vancouver, ISO, and other styles
17

Ran, Dan, Wei-Jong Shia, Miao-Chia Lo, Jun-Bao Fan, David A. Knorr, Patrick I. Ferrell, Zhaohui Ye, et al. "RUNX1a enhances hematopoietic lineage commitment from human embryonic stem cells and inducible pluripotent stem cells." Blood 121, no. 15 (April 11, 2013): 2882–90. http://dx.doi.org/10.1182/blood-2012-08-451641.

Full text
Abstract:
Abstract Advancements in human pluripotent stem cell (hPSC) research have potential to revolutionize therapeutic transplantation. It has been demonstrated that transcription factors may play key roles in regulating maintenance, expansion, and differentiation of hPSCs. In addition to its regulatory functions in hematopoiesis and blood-related disorders, the transcription factor RUNX1 is also required for the formation of definitive blood stem cells. In this study, we demonstrated that expression of endogenous RUNX1a, an isoform of RUNX1, parallels with lineage commitment and hematopoietic emergence from hPSCs, including both human embryonic stem cells and inducible pluripotent stem cells. In a defined hematopoietic differentiation system, ectopic expression of RUNX1a facilitates emergence of hematopoietic progenitor cells (HPCs) and positively regulates expression of mesoderm and hematopoietic differentiation-related factors, including Brachyury, KDR, SCL, GATA2, and PU.1. HPCs derived from RUNX1a hPSCs show enhanced expansion ability, and the ex vivo–expanded cells are capable of differentiating into multiple lineages. Expression of RUNX1a in embryoid bodies (EBs) promotes definitive hematopoiesis that generates erythrocytes with β-globin production. Moreover, HPCs generated from RUNX1a EBs possess ≥9-week repopulation ability and show multilineage hematopoietic reconstitution in vivo. Together, our results suggest that RUNX1a facilitates the process of producing therapeutic HPCs from hPSCs.
APA, Harvard, Vancouver, ISO, and other styles
18

Wheeler, MB. "Development and validation of swine embryonic stem cells: a review." Reproduction, Fertility and Development 6, no. 5 (1994): 563. http://dx.doi.org/10.1071/rd9940563.

Full text
Abstract:
The establishment of embryonic cell lines from swine should be useful for studies of cell differentiation, developmental gene regulation and the production of transgenics. This paper summarizes the establishment of porcine (Sus scrofa) embryonic stem (ES) cell lines from preimplantation blastocysts and their ability to develop into normal chimaeras. ES cells can spontaneously differentiate into cystic embryoid bodies with ectodermal, endodermal, and mesodermal cell types. Further, culture of ES cells to confluence or induction of differentiation with retinoic acid or dimethylsulfoxide results in morphological differentiation into fibroblasts, adipocytes, and epithelial, neuronal, and muscle cells. These ES cells have a normal diploid complement of 38 chromosomes. Scanning electron microscopy of the ES cells reveals a rounded or polygonal, epithelial-like cell with numerous microvilli. The differentiation of these embryonic cell lines into several cell types indicates a pluripotent cell. Furthermore, chimaeric swine have been successfully produced using such ES cells.
APA, Harvard, Vancouver, ISO, and other styles
19

Zeng, X. "Dopaminergic Differentiation of Human Embryonic Stem Cells." Stem Cells 22, no. 6 (November 1, 2004): 925–40. http://dx.doi.org/10.1634/stemcells.22-6-925.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Dale, Tina P., Shazia Mazher, William R. Webb, Jing Zhou, Nicola Maffulli, Guo-Qiang Chen, Alicia J. El Haj, and Nicholas R. Forsyth. "Tenogenic Differentiation of Human Embryonic Stem Cells." Tissue Engineering Part A 24, no. 5-6 (March 2018): 361–68. http://dx.doi.org/10.1089/ten.tea.2017.0017.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Stavridis, M. P., and A. G. Smith. "Neural differentiation of mouse embryonic stem cells." Biochemical Society Transactions 31, no. 1 (February 1, 2003): 45–49. http://dx.doi.org/10.1042/bst0310045.

Full text
Abstract:
Pluripotent embryonic stem cells can give rise to neuroectodermal derivatives in culture. This potential could be harnessed to generate neurons and glia for cell-replacement therapies in the central nervous system and for use in drug discovery. However, current methods of neural differentiation are empirical and relatively innefficient. Here, we review these methodologies and present new tools for quantification, analysis and manipulation of embryonic stem cell neural determination.
APA, Harvard, Vancouver, ISO, and other styles
22

Liu, Yinyin, Haibo Zhao, Liang Liang, Peilei Fan, Yujia Zhao, Jinling Feng, Ying Zhang, Yang Gao, and Zhengsheng Shen. "Pluripotency and differentiation of embryonic stem cells." E3S Web of Conferences 185 (2020): 04034. http://dx.doi.org/10.1051/e3sconf/202018504034.

Full text
Abstract:
Mouse embryonic stem (ES) cells derive from the inner cell mass of an early embryo called blastocyst, making them promising resource for regenerative medicine. They possess two unique properties: self-renewal and pluripotency. Different ways can be used to assess which extracellular signal and factor inside ES cells has an impact on the pluripotency of ES cells. Nowadays, many extracellular signals and transcription factors have been identified, such as extracellular signals like LIF and transcription factors like Oct4. Studying the mechanism and function of these factors offers great insight and advance our understanding of pluripotency and self-renewal and thus shed light on regenerative medicine.
APA, Harvard, Vancouver, ISO, and other styles
23

Hanley, E., J. L. Lauer, B. K. Micales, G. E. Lyons, and J. L. Shohet. "Surface-directed differentiation of embryonic stem cells." Applied Physics Letters 92, no. 19 (May 12, 2008): 193902. http://dx.doi.org/10.1063/1.2929387.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Meganathan, Kesavan, Smita Jagatp, Vilas Wagh, John Antonydas Gaspar, Johannes Winkler, Jürgen Hescheler, and Agapios Sachinidis. "Embryonic stem cells differentiation substantiates thalidomide teratogenicity." Toxicology Letters 211 (June 2012): S75. http://dx.doi.org/10.1016/j.toxlet.2012.03.289.

Full text
APA, Harvard, Vancouver, ISO, and other styles
25

Jones, Elizabeth A., David Tosh, David I. Wilson, Susan Lindsay, and Lesley M. Forrester. "Hepatic Differentiation of Murine Embryonic Stem Cells." Clinical Science 103, s47 (July 1, 2002): 37P. http://dx.doi.org/10.1042/cs103037pa.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Filippi, Marie-Dominique, Françoise Porteu, Françoise Le Pesteur, Philippe Rameau, Maria Manuela Nogueira, Najet Debili, William Vainchenker, Frederic J. de Sauvage, Anne Dubart Kupperschmitt, and Françoise Sainteny. "Embryonic stem cell differentiation to hematopoietic cells." Experimental Hematology 28, no. 12 (December 2000): 1363–72. http://dx.doi.org/10.1016/s0301-472x(00)00549-x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Jones, E. "Hepatic differentiation of murine embryonic stem cells." Journal of Hepatology 34 (April 2001): 27–28. http://dx.doi.org/10.1016/s0168-8278(01)80082-9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Jones, Elizabeth A., David Tosh, Susan Lindsay, David I. Wilson, and Lesley M. Forrester. "Hepatic differentiation of murine embryonic stem cells." Journal of Hepatology 34 (April 2001): 27–28. http://dx.doi.org/10.1016/s0168-8278(01)80957-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Keller, Gordon M. "In vitro differentiation of embryonic stem cells." Current Opinion in Cell Biology 7, no. 6 (January 1995): 862–69. http://dx.doi.org/10.1016/0955-0674(95)80071-9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
30

THOMSON, JAMES A., VIVIENNE S. MARSHALL, and JOHN Q. TROJANOWSKI. "Neural differentiation of rhesus embryonic stem cells." APMIS 106, no. 1-6 (January 1998): 149–57. http://dx.doi.org/10.1111/j.1699-0463.1998.tb01330.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
31

Smith, Austin G. "Culture and differentiation of embryonic stem cells." Journal of Tissue Culture Methods 13, no. 2 (June 1991): 89–94. http://dx.doi.org/10.1007/bf01666137.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Wang, Xiuli, and Kaiming Ye. "Directed pancreatic differentiation from embryonic stem cells." Journal of Biotechnology 136 (October 2008): S127. http://dx.doi.org/10.1016/j.jbiotec.2008.07.267.

Full text
APA, Harvard, Vancouver, ISO, and other styles
33

Dhara, Sujoy K., and Steven L. Stice. "Neural differentiation of human embryonic stem cells." Journal of Cellular Biochemistry 105, no. 3 (October 15, 2008): 633–40. http://dx.doi.org/10.1002/jcb.21891.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Chen, Xuesong, and Fanyi Zeng. "Directed hepatic differentiation from embryonic stem cells." Protein & Cell 2, no. 3 (March 2011): 180–88. http://dx.doi.org/10.1007/s13238-011-1023-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Tiruthani, Karthik, Prasenjit Sarkar, and Balaji Rao. "Trophoblast differentiation of human embryonic stem cells." Biotechnology Journal 8, no. 4 (January 17, 2013): 421–33. http://dx.doi.org/10.1002/biot.201200203.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Jones, Elizabeth A., David Tosh, David I. Wilson, Susan Lindsay, and Lesley M. Forrester. "Hepatic Differentiation of Murine Embryonic Stem Cells." Experimental Cell Research 272, no. 1 (January 2002): 15–22. http://dx.doi.org/10.1006/excr.2001.5396.

Full text
APA, Harvard, Vancouver, ISO, and other styles
37

Yasuda, Satoshi, Tetsuya Hasegawa, Tetsuji Hosono, Mitsutoshi Satoh, Kei Watanabe, Kageyoshi Ono, Shunichi Shimizu, et al. "AW551984: a novel regulator of cardiomyogenesis in pluripotent embryonic cells." Biochemical Journal 437, no. 2 (June 28, 2011): 345–55. http://dx.doi.org/10.1042/bj20110520.

Full text
Abstract:
An understanding of the mechanism that regulates the cardiac differentiation of pluripotent stem cells is necessary for the effective generation and expansion of cardiomyocytes as cell therapy products. In the present study, we have identified genes that modulate the cardiac differentiation of pluripotent embryonic cells. We isolated P19CL6 cell sublines that possess distinct properties in cardiomyogenesis and extracted 24 CMR (cardiomyogenesis-related candidate) genes correlated with cardiomyogenesis using a transcriptome analysis. Knockdown of the CMR genes by RNAi (RNA interference) revealed that 18 genes influence spontaneous contraction or transcript levels of cardiac marker genes in EC (embryonal carcinoma) cells. We also performed knockdown of the CMR genes in mouse ES (embryonic stem) cells and induced in vitro cardiac differentiation. Three CMR genes, AW551984, 2810405K02Rik (RIKEN cDNA 2810405K02 gene) and Cd302 (CD302 antigen), modulated the cardiac differentiation of both EC cells and ES cells. Depletion of AW551984 attenuated the expression of the early cardiac transcription factor Nkx2.5 (NK2 transcription factor related locus 5) without affecting transcript levels of pluripotency and early mesoderm marker genes during ES cell differentiation. Activation of Wnt/β-catenin signalling enhanced the expression of both AW551984 and Nkx2.5 in ES cells during embryoid body formation. Our findings indicate that AW551984 is a novel regulator of cardiomyogenesis from pluripotent embryonic cells, which links Wnt/β-catenin signalling to Nkx2.5 expression.
APA, Harvard, Vancouver, ISO, and other styles
38

Azab, Azab. "Stem Cells: Insights into Niche, Classification, Identification, Characterization, Mechanisms of Regeneration by Using Stem Cells, and Applications in Joint Disease Remedy." Biotechnology and Bioprocessing 2, no. 1 (February 1, 2021): 01–07. http://dx.doi.org/10.31579/2766-2314/024.

Full text
Abstract:
Background: Stem cell therapy has attracted much interest in the 21st century, not only because of the controversy surrounding the ethics involving pluripotent stem cells, but their potential for clinical use. Objectives: The present review highlights the stem cells niche, types, identification, and characterization, mechanisms of regeneration by using stem cells, and applications in joint disease remedy. Stem cells could be well differentiated cells with the potential to display different cell types depending on the host niche. Niche is defined as the cellular microenvironment providing support and stimuli to control the properties of stem cells. It consists of signaling molecules, inter-cell contacts and interaction between stem cells and their extracellular matrix neighbors. Stem cells are classified according to their sources into two main types, the embryonic and non-embryonic. Embryonic stem cells are pluripotent and can differentiate into all germ layers. Non-embryonic stem cells can be sub-classified into fetal stem cells and adult stem cells. Cultured cells can be made to differentiate into exclusive lineages by providing selective media components that can be identified by histochemical staining and quantified by quantitative Real-time polymerase chain reaction. Mesenchymal stem cells (MSCs) can be identified based on the expression of specific proteins called surface antigen phenotype of mesenchymal stem cell markers. MSCs secrete a variety of interleukins, several neurotrophic factors, many cytokines, and growth factors. These secreted bioactive factors have both paracrine and autocrine effects, which are anti-fibrotic and anti-apoptotic, as well as enhance angiogenesis. Furthermore, they stimulate mitosis and differentiation of tissue-intrinsic reparative stem cells. Systemic MSC transplantation can engraft to an injured tissue and promote wound healing through differentiation, and proliferation in synergy with hematopoietic stem cells. MSCs have been shown to express a variety of chemokines and chemokine receptors and can home to sites of inflammation by migrating towards injury or inflammatory chemokines and cytokines. MSCs are proven to have immunomodulatory properties that are among the most intriguing aspects of their biology. The immunosuppressive properties of MSCs inhibit the immune response of naive and memory T cells in a mixed lymphocyte culture and induce mitogen. The systemic infusion of MSCs can be used in immunosuppressive therapy of various disorders. MSCs have become an alternative source of cells that can be drawn from several these cells have been used as treatment to repair cartilage defects at early stages sources. Using the MSCs and directing them into chondrogenic differentiation might lead to the formation of higher quality cartilage, which has a great composition of hyaline, adequate structural reorganization and therefore improved biomechanical properties. Conclusion: It can be concluded that stem cells are classified according to their sources into two main types, the embryonic and non-embryonic. Embryonic stem cells are pluripotent and can differentiate into all germ layers. Non-embryonic stem cells can be sub-classified into fetal stem cells and adult stem cells. MSCs secrete bioactive factors that are anti-fibrotic and anti-apoptotic, as well as enhance angiogenesis. The systemic infusion of MSCs can be used in immunosuppressive therapy of various disorders. These cells have been used as treatment to repair cartilage defects at early stages.
APA, Harvard, Vancouver, ISO, and other styles
39

Perlingeiro, Rita C. R., Michael Kyba, and George Q. Daley. "Clonal analysis of differentiating embryonic stem cells reveals a hematopoietic progenitor with primitive erythroid and adult lymphoid-myeloid potential." Development 128, no. 22 (November 15, 2001): 4597–604. http://dx.doi.org/10.1242/dev.128.22.4597.

Full text
Abstract:
Embryonic stem (ES) cells differentiate into multiple hematopoietic lineages during embryoid body formation in vitro, but to date, an ES-derived hematopoietic stem cell has not been identified and subjected to clonal analysis in a manner comparable with hematopoietic stem cells from adult bone marrow. As the chronic myeloid leukemia-associated BCR/ABL oncogene endows the adult hematopoietic stem cell with clonal dominance without inhibiting pluripotent lymphoid and myeloid differentiation, we have used BCR/ABL as a tool to enable engraftment and clonal analysis. We show that embryoid body-derived hematopoietic progenitors expressing BCR/ABL maintain a primitive hematopoietic blast stage of differentiation and generate only primitive erythroid cell types in vitro. These cells can be cloned, and when injected into irradiated adult mice, they differentiate into multiple myeloid cell types as well as T and B lymphocytes. While the injected cells express embryonic (β-H1) globin, donor-derived erythroid cells in the recipient express only adult (β-major) globin, suggesting that these cells undergo globin gene switching and developmental maturation in vivo. These data demonstrate that an embryonic hematopoietic stem cell arises in vitro during ES cell differentiation that constitutes a common progenitor for embryonic erythroid and definitive lymphoid-myeloid hematopoiesis.
APA, Harvard, Vancouver, ISO, and other styles
40

Mulvey, Claire M., Christian Schröter, Laurent Gatto, Duygu Dikicioglu, Isik Baris Fidaner, Andy Christoforou, Michael J. Deery, et al. "Dynamic Proteomic Profiling of Extra-Embryonic Endoderm Differentiation in Mouse Embryonic Stem Cells." STEM CELLS 33, no. 9 (June 23, 2015): 2712–25. http://dx.doi.org/10.1002/stem.2067.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

Olivier, Emmanuel N., Anne C. Rybicki, and Eric E. Bouhassira. "Differentiation of Human Embryonic Stem Cells into Bipotent Mesenchymal Stem Cells." Stem Cells 24, no. 8 (August 2006): 1914–22. http://dx.doi.org/10.1634/stemcells.2005-0648.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Cai, Chunyu, and Laura Grabel. "Directing the differentiation of embryonic stem cells to neural stem cells." Developmental Dynamics 236, no. 12 (2007): 3255–66. http://dx.doi.org/10.1002/dvdy.21306.

Full text
APA, Harvard, Vancouver, ISO, and other styles
43

Levenberg, Shulamit, Janet Zoldan, Yaara Basevitch, and Robert Langer. "Endothelial potential of human embryonic stem cells." Blood 110, no. 3 (August 1, 2007): 806–14. http://dx.doi.org/10.1182/blood-2006-08-019190.

Full text
Abstract:
Abstract Growing interest in using endothelial cells for therapeutic purposes has led to exploring human embryonic stem cells as a potential source for endothelial progenitor cells. Embryonic stem cells are advantageous when compared with other endothelial cell origins, due to their high proliferation capability, pluripotency, and low immunogenity. However, there are many challenges and obstacles to overcome before the vision of using embryonic endothelial progenitor cells in the clinic can be realized. Among these obstacles is the development of a productive method of isolating endothelial cells from human embryonic stem cells and elucidating their differentiation pathway. This review will focus on the endothelial potential of human embryonic stem cells that is described in current studies, with respect to the differentiation of human embryonic stem cells to endothelial cells, their isolation, and their characterization.
APA, Harvard, Vancouver, ISO, and other styles
44

Chang, Kai-Hsin, Angelique M. Nelson, Hua Cao, Linlin Wang, Betty Nakamoto, Carol B. Ware, and Thalia Papayannopoulou. "Definitive-like erythroid cells derived from human embryonic stem cells coexpress high levels of embryonic and fetal globins with little or no adult globin." Blood 108, no. 5 (September 1, 2006): 1515–23. http://dx.doi.org/10.1182/blood-2005-11-011874.

Full text
Abstract:
Human embryonic stem cells are a promising tool to study events associated with the earliest ontogenetic stages of hematopoiesis. We describe the generation of erythroid cells from hES (H1) by subsequent processing of cells present at early and late stages of embryoid body (EB) differentiation. Kinetics of hematopoietic marker emergence suggest that CD45+ hematopoiesis peaks at late D14EB differentiation stages, although low-level CD45- erythroid differentiation can be seen before that stage. By morphologic criteria, hES-derived erythroid cells were of definitive type, but these cells both at mRNA and protein levels coexpressed high levels of embryonic (ϵ) and fetal (γ) globins, with little or no adult globin (β). This globin expression pattern was not altered by the presence or absence of fetal bovine serum, vascular endothelial growth factor, Flt3-L, or coculture with OP-9 during erythroid differentiation and was not culture time dependent. The coexpression of both embryonic and fetal globins by definitive-type erythroid cells does not faithfully mimic either yolk sac embryonic or their fetal liver counterparts. Nevertheless, the high frequency of erythroid cells coexpressing embryonic and fetal globin generated from embryonic stem cells can serve as an invaluable tool to further explore molecular mechanisms.
APA, Harvard, Vancouver, ISO, and other styles
45

Annerén, Cecilia. "Tyrosine kinase signalling in embryonic stem cells." Clinical Science 115, no. 2 (June 12, 2008): 43–55. http://dx.doi.org/10.1042/cs20070388.

Full text
Abstract:
Pluripotent ES (embryonic stem) cells can be expanded in culture and induced to differentiate into a wide range of cell types. Self-renewal of ES cells involves proliferation with concomitant suppression of differentiation. Some critical and conserved pathways regulating self-renewal in both human and mouse ES cells have been identified, but there is also evidence suggesting significant species differences. Cytoplasmic and receptor tyrosine kinases play important roles in proliferation, survival, self-renewal and differentiation in stem, progenitor and adult cells. The present review focuses on the role of tyrosine kinase signalling for maintenance of the undifferentiated state, proliferation, survival and early differentiation of ES cells.
APA, Harvard, Vancouver, ISO, and other styles
46

Estiri, Hajar, Ali Fallah, and Mansoure Movahedin. "Mouse Embryonic Stem Cells Differentiation to Neuron-like Cells." Neuroscience Journal of Shefaye Khatam 1, no. 4 (December 1, 2013): 9–16. http://dx.doi.org/10.18869/acadpub.shefa.1.4.9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Derebail, Suchitra, Casthri Krishnamurthy, Ong Hong Boon, Ang Kailin, Nur Amilia Bte M. Isa, Nur Ayuni Bte Hassan Jaya, and Orr Hui Min. "REVIEW." Asia-Pacific Biotech News 18, no. 01 (January 2014): 47–51. http://dx.doi.org/10.1142/s0219030314000068.

Full text
APA, Harvard, Vancouver, ISO, and other styles
48

Ulmschneider, Bryne, Bree K. Grillo-Hill, Marimar Benitez, Dinara R. Azimova, Diane L. Barber, and Todd G. Nystul. "Increased intracellular pH is necessary for adult epithelial and embryonic stem cell differentiation." Journal of Cell Biology 215, no. 3 (November 7, 2016): 345–55. http://dx.doi.org/10.1083/jcb.201606042.

Full text
Abstract:
Despite extensive knowledge about the transcriptional regulation of stem cell differentiation, less is known about the role of dynamic cytosolic cues. We report that an increase in intracellular pH (pHi) is necessary for the efficient differentiation of Drosophila adult follicle stem cells (FSCs) and mouse embryonic stem cells (mESCs). We show that pHi increases with differentiation from FSCs to prefollicle cells (pFCs) and follicle cells. Loss of the Drosophila Na+–H+ exchanger DNhe2 lowers pHi in differentiating cells, impairs pFC differentiation, disrupts germarium morphology, and decreases fecundity. In contrast, increasing pHi promotes excess pFC cell differentiation toward a polar/stalk cell fate through suppressing Hedgehog pathway activity. Increased pHi also occurs with mESC differentiation and, when prevented, attenuates spontaneous differentiation of naive cells, as determined by expression of microRNA clusters and stage-specific markers. Our findings reveal a previously unrecognized role of pHi dynamics for the differentiation of two distinct types of stem cell lineages, which opens new directions for understanding conserved regulatory mechanisms.
APA, Harvard, Vancouver, ISO, and other styles
49

Dvorakova, Dana, Stanislava Koskova, Martina Vodinska, Ales Hampl, Jiri Mayer, and Petr Dvorak. "Fibroblast Growth Factor Receptors in Human Embryonic Stem Cells." Blood 104, no. 11 (November 16, 2004): 4168. http://dx.doi.org/10.1182/blood.v104.11.4168.4168.

Full text
Abstract:
Abstract Signals via fibroblast growth factor receptors (FGFRs) are involved in mesoderm induction events and may be also critical for early hematopoietic specification and proliferation of the hemangioblast. In vitro differentiated embryonic stem cells represent excellent system for the study of early hematopoietic commitment, particularly for understanding signals regulating the onset of hematopoietic differentiation. We have used human embryonic stem cells (hESCs) to study the expression of FGFR1, 2, 3, and 4 in undifferentiated cells and their differentiated progeny. Culturing hESCs i/ in high densities (protocol 1), ii/ without feeder layer of mouse embryonal fibroblasts and basic fibroblast growth factor (protocol 2), and iii/ in three-dimensional aggregates called embryoid bodies (protocol 3), was used to induce the differentiation. To achieve more directed and homogenous differentiation feeder-free hESCs were first subjected to the aggregation step (formation of embryoid bodies) that resembles the gastrulation process. This was followed by differentiation in monolayer in the presence of basic fibroblast growth factor (protocol 4). Such two-step differentiation protocol (5 + 10 days) was shown to activate ectodermal and mesodermal genes and form ectodermal and mesodermal cells (Schuldiner et al., PNAS97:11307, 2000). The gene expression levels for all FGFRs were determined by quantitative real-time RT-PCR. Real-time RT-PCR results were normalized by comparison to the expression of ABL gene. We revealed that undifferentiated hESCs that were cultured with feeder cells and in low density express all four FGFRs in the following pattern: FGFR1 is highly expressed and dominant; FGFR3 is also strongly expressed; FGFR4 shows lower expression; and FGFR2 is only weakly expressed. This expression pattern was changed when hESCs grew and started to differentiate in high densities (protocol 1) or have initiated differentiation either by feeder cells and basic fibroblast growth factor withdrawal or by aggregation step (protocol 2 and 3). Two-fold upregulation of FGFR1 and FGFR4, and downregulation of FGFR3 characterize such changed expression pattern. Notably, the expression levels for all four FGFRs were increased when hESCc were subjected to the two-step differentiation protocol (protocol 4). Compared to the undifferentiated hESCs, FGFR1 and 4 exhibited 7-fold increase, and FGFR2 and 3 were found to be upregulated more than twice. In summary our results show that the expression of FGFRs tightly follows changing culture conditions that may direct hESCs to differentiate. Furthermore, strong upregulation of FGFR1 and 4 in prospective hESC-derived mesodermal cells suggests their involvement in the earliest stages of hematopoiesis. This research was supported in part by the Grant Agency of the Czech Republic (301/03/1122), Ministry of Health (MZ 00065269705), Ministry of Education, Youth, and Sports (MSM 432100001, LN 00A065), and Academy of Sciences of the Czech Republic (AV 0Z5039906).
APA, Harvard, Vancouver, ISO, and other styles
50

Narita, N., M. Bielinska, and D. B. Wilson. "Cardiomyocyte differentiation by GATA-4-deficient embryonic stem cells." Development 124, no. 19 (October 1, 1997): 3755–64. http://dx.doi.org/10.1242/dev.124.19.3755.

Full text
Abstract:
In situ hybridization studies, promoter analyses and antisense RNA experiments have implicated transcription factor GATA-4 in the regulation of cardiomyocyte differentiation. In this study, we utilized Gata4−/− embryonic stem (ES) cells to determine whether this transcription factor is essential for cardiomyocyte lineage commitment. First, we assessed the ability of Gata4−/− ES cells form cardiomyocytes during in vitro differentiation of embryoid bodies. Contracting cardiomyocytes were seen in both wild-type and Gata4−/− embryoid bodies, although cardiomyocytes were observed more often in wild type than in mutant embryoid bodies. Electron microscopy of cardiomyocytes in the Gata4−/− embryoid bodies revealed the presence of sarcomeres and junctional complexes, while immunofluorescence confirmed the presence of cardiac myosin. To assess the capacity of Gata4−/− ES cells to differentiate into cardiomyocytes in vivo, we prepared and analyzed chimeric mice. Gata4−/− ES cells were injected into 8-cell-stage embryos derived from ROSA26 mice, a transgenic line that expresses beta-galactosidase in all cell types. Chimeric embryos were stained with X-gal to discriminate ES cell- and host-derived tissue. Gata4−/− ES cells contributed to endocardium, myocardium and epicardium. In situ hybridization showed that myocardium derived from Gata4−/− ES cells expressed several cardiac-specific transcripts, including cardiac alpha-myosin heavy chain, troponin C, myosin light chain-2v, Nkx-2.5/Csx, dHAND, eHAND and GATA-6. Taken together these results indicate that GATA-4 is not essential for terminal differentiation of cardiomyocytes and suggest that additional GATA-binding proteins known to be in cardiac tissue, such as GATA-5 or GATA-6, may compensate for a lack of GATA-4.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography