Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-06-12T18:51:53.664Z Has data issue: false hasContentIssue false

35 - MicroRNAs in embryonic stem cell differentiation and prediction of their targets

from VI - MicroRNAs in stem cell development

Published online by Cambridge University Press:  22 August 2009

Yvonne Tay
Affiliation:
Stem Cell and Developmental Biology Genome Institute of Singapore 60 Biopolis Street, #02-01, Genome Singapore 138672
Andrew M. Thomson
Affiliation:
Stem Cell and Developmental Biology Genome Institute of Singapore 60 Biopolis Street, #02-01, Genome Singapore 138672
Bing Lim
Affiliation:
Stem Cell and Developmental Biology Genome Institute of Singapore 60 Biopolis Street, #02-01, Genome Singapore 138672
Get access

Summary

Introduction

Embryonic stem cells (ESCs) exhibit the capacity for unlimited self-renewal and the ability to differentiate into multiple cell lineages, and are termed pluripotent (Smith, 2001; Czyz et al., 2003). The study of mammalian ESCs may facilitate understanding of early developmental events and contribute to the advance of cell-based regenerative medicine (Loebel at al., 2003). ESCs are isolated from the inner cell mass (ICM) of the pre-implantation embryo (Evans and Kaufman, 1981) and share many properties with pluripotent cells from the ICM (Figure 35.1). These include the expression of key regulators of pluripotency and self-renewal, Oct4, Sox2 and Nanog, and the capacity to differentiate to all cell types of the embryo, including mesoderm, ectoderm and endoderm (Orkin, 2005). Oct4 is activated at the four cell stage of mouse pre-implantation development and is essential for the formation of the ICM (Chew et al., 2005). Increased Oct4 levels in mouse ESCs (mESCs) result in differentiation to primitive endoderm and mesoderm, whereas a reduction leads to differentiation to trophectoderm (Niwa et al., 2000). Co-regulation of genes by Oct4 and Sox2 appears to be essential in maintaining self-renewal of ESCs (Chew et al., 2005). Nanog acts in concert with Oct4 and Sox2, where Nanog overexpression negates the requirement for leukemia-inhibitory factor (LIF)-activated signaling by mESCs for self-renewal (Mitsui et al., 2003). Nanog knockout results in differentiation of mESCs into parietal/visceral endoderm (Mitsui et al., 2003).

Type
Chapter
Information
MicroRNAs
From Basic Science to Disease Biology
, pp. 476 - 488
Publisher: Cambridge University Press
Print publication year: 2007

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Balmer, L., Beveridge, D. J., Jazayeri, J. A.et al. (2001). Identification of a novel AU-rich element in the 3′ UTR of epidermal growth factor receptor mRNA that is the target for regulated RNA-binding proteins. Molecular and Cellular Biology, 21, 2070–2084.Google Scholar
Boyer, L. A., Lee, T. I., Cole, M. F., Johnstone, S. E. and Levine, S. S. (2005). Core transcriptional regulatory circuitry in human embryonic stem cells. Cell, 122, 947–956.Google Scholar
Brennecke, J., Hipfner, D. R., Stark, A., Russell, R. B. and Cohen, S. M. (2003). bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell, 113, 25–36.CrossRefGoogle Scholar
Chen, C. Z., Li, L., Lodish, H. F. and Bartel, D. P. (2004). MicroRNAs modulate hematopoietic lineage differentiation. Science, 303, 83–86.CrossRefGoogle Scholar
Cheng, L., Tavazole, M. and Doetsch, F. (2005). Stem cells: from epigenetics to microRNAs. Neuron, 46, 363–367.CrossRefGoogle Scholar
Chew, J. L., Loh, Y. H., Zhang, W.et al. (2005). Reciprocal transcriptional regulation of Pou5f1 and Sox2 via the Oct4/Sox2 complex in embryonic stem cells. Molecular and Cellular Biology, 25, 6031–6046.Google Scholar
Czyz, J., Wiese, C., Rolletschek, A.et al. (2003). Potential of embryonic and adult stem cells in vitro. Biological Chemistry, 384, 1391–1409.CrossRefGoogle Scholar
Enright, A. J., John, B., Gaul, U.et al. (2003). MicroRNA targets in Drosophila. Genome Biology, 5, R1.Google Scholar
Esau, C., Kang, X., Peralta, E.et al. (2004). MicroRNA-143 regulates adipocyte differentiation. Journal of Biological Chemistry, 279, 52361–52365.CrossRefGoogle Scholar
Evans, M. J. and Kaufman, M. H. (1981). Establishment in culture of pluripotential cells from mouse embryos. Nature, 292, 154–156.CrossRefGoogle Scholar
Giles, K. M., Daly, J. M., Beveridge, D. J.et al. (2003). The 3′ untranslated region of p21WAF1 mRNA is a composite cis-acting sequence bound by RNA-binding proteins from breast cancer cells, including HuR and poly(C)-binding protein. Journal of Biological Chemistry, 278, 2937–2946.CrossRefGoogle Scholar
Giraldez, A. J., Cinalli, R. M., Glasner, M. E.et al. (2005). MicroRNAs regulate brain morphogenesis in zebrafish. Science, 308, 833–838.CrossRefGoogle Scholar
Gore, A. V., Maegawa, S., Cheong, A.et al. (2005). The zebrafish dorsal axis is apparent at the four-cell stage. Nature, 438, 1030–1035.CrossRefGoogle Scholar
Gregory, R. I., Chendrimuka, T. P., Cooch, N. and Shiekhattar, R. (2005). Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell, 123, 631–640.CrossRefGoogle Scholar
He, L., Thomson, J. M., Hemann, M. T.et al. (2005). A microRNA polycistron as a potential human oncogene. Nature, 435, 828–33.Google Scholar
Hollams, E. M., Giles, K. M., Thomson, A. M. and Leedman, P. J. (2002). mRNA stability and the control of gene expression: implications for human disease. Neurochemical Research, 27, 957–980.CrossRefGoogle Scholar
Houbaviy, H. B., Murray, M. F. and Sharp, P. A. (2003). Embryonic stem cell-specific microRNAs. Developmental Cell, 5, 351–358.Google Scholar
Houbaviy, H. B., Dennis, L., Jaenisch, R. and Sharp, P. A. (2005). Characterization of a highly variable eutherian microRNA gene. RNA, 11, 1–13.Google Scholar
Jin, P., Zarnescu, D. C., Ceman, S.et al. (2004). Biochemical and genetic interaction between the fragile X mental retardation protein and the microRNA pathway. Nature Neuroscience, 7, 113–117.CrossRefGoogle Scholar
John, B., Enright, A. J., Aravin, A.et al. (2004). Human microRNA targets. Public Library of Science Biology, 2, e363.Google Scholar
Kanellopoulou, C., Muljo, S. A., Kung, A. L.et al. (2005). Dicer-deficient mouse embryonic stem cells are defective in differentiation and centromeric silencing. Genes & Development, 19, 489–501.CrossRefGoogle Scholar
Kim, K. (2005). Personal communication from Keystone Symposia on Stem Cells, Senescence and Cancer, Singapore, 2005.
Kiriakidou, M., Nelson, P. T., Kouranov, A.et al. (2004). A combined computational-experimental approach predicts human microRNA targets. Genes & Development, 18, 1165–1178.CrossRefGoogle Scholar
Krek, A., Grun, D., Poy, M. N.et al. (2005). Combinatorial microRNA target predictions. Nature Genetics, 37, 495–500.CrossRefGoogle Scholar
Krichevsky, A., Sonntag, K., Isacson, O. and Kosik, K. S. (2005). Specific microRNAs modulate ES cell-derived neurogenesis. Stem Cells, Dec 15 (Epub ahead of print).Google Scholar
Lee, Y. S., Kim, H. K., Chung, S., Kim, K. and Dutta, A. (2005). Depletion of human micro-RNA miR-125b reveals that it is critical for the proliferation of differentiated cells but not for the downregulation of putative targets during differentiation. Journal of Biological Chemistry, 280, 16 635–16 641.Google Scholar
Lewis, B. P., Shih, I. H., Jones-Rhoades, M. W.et al. (2003). Prediction of mammalian microRNA targets. Cell, 115, 787–798.CrossRefGoogle Scholar
Liu, J., Rivas, F. V., Wohlschlegel, J.et al. (2005a). A role for the P-body component GW182 in microRNA function. Nature Cell Biology, Nov 13 (Epub ahead of print).Google Scholar
Liu, J., Valencia-Sanchez, M. A., Hannon, G. J. and Parker, R. (2005b). MicroRNA-dependent localization of targeted mRNAs to mammalian P-bodies. Nature Cell Biology, 7, 719–723.Google Scholar
Loebel, D. A., Watson, C. M., Young, R. A. and Tam, P. P. (2003). Lineage choice and differentiation in mouse embryos and embryonic stem cells. Developmental Biology, 264, 1–14.CrossRefGoogle Scholar
Loh, Y. H., Wu, Q., Chew, J. L.et al. (2006). The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nature Genetics, 38, 431–440.CrossRefGoogle Scholar
Lu, J., Getz, G., Miska, E. A.et al. (2005). MicroRNA expression profiles classify human cancers. Nature, 435, 834–838.CrossRefGoogle Scholar
Mansfield, J. H., Harfe, B. D., Nissen, R.et al. (2004). MicroRNA-responsive “sensor” transgenes uncover Hox-like and other developmentally regulated patterns of vertebrate microRNA expression. Nature Genetics, 36, 1079–1083.CrossRefGoogle Scholar
Marx, J. (2005). Molecular biology. P-bodies mark the spot for controlling protein production. Science, 310, 764–765.CrossRefGoogle Scholar
McManus, M. T. (2003). MicroRNAs and cancer. Seminars in Cancer Biology, 13, 253–258.CrossRefGoogle Scholar
Meister, G., Landthaler, M., Patkaniowska, A.et al. (2004). Human Argonaute2 mediates RNA cleavage targeted by miRNAs and siRNAs. Molecular Cell, 15, 185–197.Google Scholar
Metzler, M., Wilda, M., Busch, K., Viehmann, S. and Borkhardt, A. (2004). High expression of precursor microRNA-155/BIC RNA in children with Burkitt lymphoma. Genes, Chromosomes & Cancer, 39, 167–169.CrossRefGoogle Scholar
Miranda, K. C., Huynh, T., Tay, Y.et al. (2006). A pattern-based method for the identification of microRNA binding sites and their corresponding heteroduplexes. Cell, 126, 1203–1217.CrossRefGoogle Scholar
Miska, E. A. (2005). How microRNAs control cell division, differentiation and death. Current Opinion in Genetics & Development, 15, 563–568.Google Scholar
Mitsui, K., Tokuzawa, Y., Itoh, H.et al. (2003). The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell, 113, 631–642.CrossRefGoogle Scholar
Muckenthaler, M., Gray, N. K. and Hentze, M. W. (1998). IRP-1 binding to ferritin mRNA prevents the recruitment of the small ribosomal subunit by the cap-binding complex eIF4F. Molecular Cell, 2, 383–388.CrossRefGoogle Scholar
Niwa, H., Miyazaki, J. and Smith, A. G. (2000). Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nature Genetics, 24, 372–376.CrossRefGoogle Scholar
Orkin, S. H. (2005). Chipping away at the embryonic stem cell network. Cell, 122, 828–830.Google Scholar
Poy, M. N., Eliasson, L., Krutzfeldt, J.et al. (2004). A pancreatic islet-specific microRNA regulates insulin secretion. Nature, 432, 226–230.Google Scholar
Proudfoot, N. J., Furger, A. and Dye, M. J. (2002). Integrating mRNA processing with transcription. Cell, 108, 501–512.CrossRefGoogle Scholar
Rand, T. A., Petersen, S., Du, F. and Wang, X. (2005). Argonaute2 cleaves the anti-guide strand of siRNA during RISC activation. Cell, 123, 621–629.CrossRefGoogle Scholar
Rehmsmeier, M., Steffen, P., Hochsmann, M. and Giegerich, R. (2004). Fast and effective prediction of microRNA/target duplexes. RNA, 10, 1507–1517.CrossRefGoogle Scholar
Rigoutsos, I. and Floratos, A. (1998). Combinatorial pattern discovery in biological sequences: The TEIRESIAS algorithm. Bioinformatics, 14, 55–67.CrossRefGoogle Scholar
Rouault, T. A. (2005). The intestinal heme transporter revealed. Cell, 122, 649–651.CrossRefGoogle Scholar
Schratt, G. M., Tuebing, F., Nigh, E. A.et al. (2006). A brain-specific microRNA regulates dendritic spine development. Nature, 439, 283–289.CrossRefGoogle Scholar
Schulman, B. R., Esquela-Kerscher, A. and Slack, F. J. (2005). Reciprocal expression of lin-41 and the microRNAs let-7 and mir-125 during mouse embryogenesis. Developmental Dynamics, 234, 1046–1054.Google Scholar
Seitz, H., Royo, H., Bortolin, M. L.et al. (2004). A large imprinted microRNA gene cluster at the mouse Dlk1-Gtl2 domain. Genome Research, 14, 1741–1748.Google Scholar
Smith, A. G. (2001). Embryo-derived stem cells: of mice and men. Annual Review of Cell and Developmental Biology, 17, 435–462.CrossRefGoogle Scholar
Stark, A., Brennecke, J., Bushati, N., Russell, R. B. and Cohen, S. M. (2005). Animal microRNAs confer robustness to gene expression and have a significant impact on 3′ UTR evolution. Cell, 123, 1133–1146.CrossRefGoogle Scholar
Suh, M. R., Lee, Y., Kim, J. Y.et al. (2004). Human embryonic stem cells express a unique set of microRNAs. Developmental Biology, 270, 488–498.Google Scholar
Takamizawa, J., Konishi, H., Yanagisawa, K.et al. (2004). Reduced expression of the let-6 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Research, 64, 3753–3756.CrossRefGoogle Scholar
Tang, F., Hajkova, P., Barton, S. C., Lao, K. and Surani, M. A. (2006). MicroRNA expression profiling of single whole embryonic stem cells. Nucleic Acids Research, 34, e9.Google Scholar
Thomson, A. M., Cahill, C. M., Cho, H.et al. (2005). The acute box cis-element in human heavy ferritin mRNA 5′-untranslated region is a unique translation enhancer that binds poly(c)-binding proteins. Journal of Biological Chemistry, 280, 30 032–30 045.Google Scholar
Thomson, J. M., Parker, J., Perou, C. M. and Hammond, S. M. (2004). A custom microarray platform for analysis of microRNA gene expression. Nature Methods, 1, 1–7.CrossRefGoogle Scholar
Voorhoeve, P. M., Sage, C. L., Schrier, M.et al. (2006). A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell, 124, 1169–1181.CrossRefGoogle Scholar
Yeap, B. B., Voon, D. C., Vivian, J. P.et al. (2002). Novel binding of HuR and poly(C)-binding protein to a conserved UC-rich motif within the 3′-untranslated region of the androgen receptor messenger RNA. Journal of Biological Chemistry, 277, 27 183–27 192.Google Scholar
Yekta, S., Shih, I. H. and Bartel, D. P. (2004). MicroRNA-directed cleavage of HOXB8 mRNA. Science, 304, 594–596.CrossRefGoogle Scholar
Zeng, Y., Yi, R. and Cullen, B. R. (2005). Recognition and cleavage of primary microRNA precursors by the nuclear processing enzyme Drosha. European Molecular Biology Organization Journal, 24, 138–148.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×