Screen of nuclear receptors for the enhanced and alternative generation of induced pluripotent stem cells

138 342 0
Screen of nuclear receptors for the enhanced and alternative generation of induced pluripotent stem cells

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

SCREEN OF NUCLEAR RECEPTORS FOR THE ENHANCED AND ALTERNATIVE GENERATION OF INDUCED PLURIPOTENT STEM CELLS DOMINIC HENG JIAN CHIEN NATIONAL UNIVERSITY OF SINGAPORE 2012 SCREEN OF NUCLEAR RECEPTORS FOR THE ENHANCED AND ALTERNATIVE GENERATION OF INDUCED PLURIPOTENT STEM CELLS DOMINIC HENG JIAN CHIEN (B.Sc (Hons), NTU) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY NUS GRADUATE SCHOOL FOR INTEGRATIVE SCIENCES AND ENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2012 Acknowledgements First and foremost, I would like to thank my supervisor, Dr Ng Huck Hui for allowing me to work in his laboratory back in 2008 I am also grateful for his invaluable guidance, insights and direction throughout my PhD stint Graduating from his laboratory has not only made me a more critical, analytical, resourceful, intelligent and independent researcher but also a more, determined and team-playing individual I would also like to thank Professor Davor Solter and Dr Thomas Lufkin, wonderful members of my thesis advisory committee They have been incredible with their advice, support and time I thank Jiang Jianming for help with the ChIP, and ChIP-sequencing experiments, Feng Bo for her help with the imaging of the teratoma tissue, Yuriy Orlov for ChIPseq analysis, Petra Kraus and Han Jianyong for relevant mouse work, Lim Seong Soo for karyotyping, and Ng Jia Hui for help with the microarray analysis I also thank Kyle M Loh for his wonderful intelligent discourse over scientific papers and concepts as well as his unfailing help to vet through all my manuscripts and papers A very big thank you goes out to Felicia Hong for her faithful love, unwavering support and countless encouragement that she has showered upon me throughout all these years She will always be in my heart forever Last but not least, I thank God for his bountiful grace for granting me the strength and wisdom to undertake this challenging PhD stint i Table of Contents Acknowledgements……………………………………………………………………i Table of Contents…………………………………………………………………….ii Summary……………………………………………………………… ………….viii List of Tables………………………………………………………………………….x List of Figures………………………………………………………… …………xi List of Symbols…………………………………………………………….……… xv Introduction……………………………………………………………………… 1.1 A plethora of cell types governed by transcriptional and epigenetic regulation… 1.2 Embryonic stem cell, the common precursor cell……………………… ……….3 1.3 Important transcription factors governing the ESC fate: Oct4, Sox2 and Nanog 1.4 The first derivations of mouse and human ESCs………………………… …… 1.5 The issues that plague the utilisation of human ESCs for regenerative medicine 1.6 Differentiation was previously conceived as an irreversible process……… …….8 1.7 Methodologies to reprogram somatic cells to a state of pluripotency……… … 10 1.7.1 Cell fusion………………………………………………………… .10 1.7.2 Somatic cell nuclear transfer………………………………………… 11 1.7.3 Cell explantation…………………………………………….………….12 ii 1.7.4 Reprogramming with transcription factors…………………… ………13 1.8 The canonical reprogramming factors………………………………….……… 14 1.8.1 Oct4 in reprogramming………………………………….…………… 14 1.8.2 Sox2 in reprogramming………………………………………… …….14 1.8.3 Klf4 in reprogramming………………………………… ……….…….15 1.8.4 c-Myc in reprogramming……………………………………………….16 1.9 Advantages of iPSCs………………………………………………… …………17 1.10 The biology of reprogramming to iPSCs………………………………….……18 1.11 The rapid development of the field of iPSC………………………… ……… 21 1.12 Various cell types from various species reprogrammed……………………….23 1.13 Various techniques to generate iPSCs………………………………………….24 1.14 Screen for reprogramming factors……………………………………… ……26 1.14.1 Initial screen for factors that could reprogram………………….…….26 1.14.2 Screen for factors that could reprogram human fibroblasts………… 29 1.14.3 Screen for factors that could enhance the generation of human iPSCs………………………………………………………………… …….31 1.14.4 Screen for factors that could replace exogenous Klf4 in reprogramming…………………………………………………… ……… 33 1.14.5 Screen of human transcription factor library………………… …….35 iii 1.14.6 Other players in reprogramming: non-coding RNAs…………………36 1.14.6.1 MicroRNAs in reprogramming…………………………… 36 1.14.6.2 LincRNAs in reprogramming…………………….…………38 1.15 Nuclear receptors……………………………………………………………… 39 Materials and Methods………………………………………………… ………42 2.1 Cell Culture…………………………………………………………… ……… 42 2.1 Transfection………………………………………………………………………42 2.3 Mouse genetics………………………………………………… ………………42 2.4 Packing of retroviruses and infection…………………………………………….43 2.5 PCR genotyping of retroviral integration into the genome………… ………… 48 2.6 Bisulphite genomic DNA sequencing…………………………………….….… 48 2.7 Embryoid body formation and in vitro differentiation……………… ………….49 2.8 Teratoma formation assay………………………………………………….…….49 2.9 TUNEL apoptosis assay………………………………………………………….50 2.10 Western blot analysis…………………………………………………… …….50 2.11 Southern blot analysis……………………………………………………… …51 2.12 Immunofluorescence and alkaline phosphatase staining……………………… 51 2.13 RNA extraction, reverse transcription and quantitative real-time PCR… ……52 2.14 G-banded karyotyping……………………………………………… …………55 iv 2.15 Microarray experiment and analysis……………………………………………55 2.16 ChIP assay………………………………………………………… ………….56 2.17 ChIP sequencing and analysis………………………………………………… 56 2.18 Gene targeting of POU5F1 locus by homologous recombination………… …57 Results ……………………………………………………………… …….…… 59 3.1 Setting up of the reprogramming assay………………………………….……… 59 3.2 Screen of nuclear receptors in reprogramming reveals Nr5a2 and Nr1i2 as enhancers…………………………………………………………………………… 59 3.3 Nr5a2 enhances both the efficiency and kinetics of reprogramming 63 3.4 Nr5a2 can replace exogenous Oct4 in reprogramming………………………… 66 3.5 Nr5a2-reprogrammed cells fulfil pluripotent assays……………………… ……71 3.6 Epigenetic and transcriptional profiles of Nr5a2-reprogrammed cells are akin to ESCs………………………………………………………………………………….74 3.7 The other nuclear receptors are unable to replace exogenous Oct4 in reprogramming………………………………………………………… ………… 78 3.8 Nr5a1, the close family member of Nr5a2 possess similar reprogramming capabilities as its counterpart…………………………………………….………… 78 3.9 Other transcription factors that bind to Oct4 regulatory region are unable to replace exogenous Oct4 in reprogramming………………………………………… 82 3.10 Nr5a2 sumoylation mutants can further enhance the efficiency of reprogramming…………………………………………………………….…………83 v 3.11 Genome wide binding analysis of Nr5a2 in mouse ESCs reveals that it shares many common target genes as its reprogramming counterparts, Sox2 and Klf4….…86 3.12 Nr5a2 works in part through Nanog in reprogramming…………………… …88 3.13 Nr5a2 works in a p53-pathway inhibition-independent fashion…………….….91 3.14 Nr5a2 does not enhance or replace exogenous Oct4 in human reprogramming…………………………………………………………………… 93 Discussion……………………………………………………………… ……… 98 4.1 The identification of more nuclear receptor factors associated with reprogramming…………………………………………………………….…………98 4.2 Nr5a2, the first factor reported that can replace exogenous Oct4 in reprogramming…………………………………………………………………….…99 4.3 The parallel importance of Nr5a2 in mouse ESCs, early embryogenesis and reprogramming……………………………………………………….…………… 101 4.4 Nr5a2 works in part through Nanog to mediate reprogramming……… …… 102 4.5 Nr5a2 additionally reprograms mouse EpiSCs besides mouse somatic cells…………………………………………………………………………….……104 4.6 Species-specific actions of Nr5a2 in reprogramming……………….………….105 4.7 Mouse ESC-like human ESCs with Nr5a2…………………… ……………….106 4.8 Possible role of Nr5a2 in transdifferentiation………………………………… 108 vi Conclusion…………………………………………………………….…………109 References……………………………………………………………………….110 vii Summary Differentiated cells are typified by their lineage restriction Nevertheless, a pluripotent state of unrestricted multilineage differentiation potential may be experimentally endowed upon differentiated cells via the process of pluripotential reprogramming in which the lineage restriction of differentiated cells is undone The resultant cells, known colloquially as induced pluripotent stem cells (iPSCs), become akin to pluripotent embryonic stem cells (ESCs) and pluripotent cells of the early embryo, thus gaining their characteristic developmental potential and other characteristics diagnostic of pluripotent cells Conferral of pluripotency upon differentiated cells is achieved by overexpressing pluripotency-associated transcription factors in these cells, such as Oct4, Sox2, Klf4, and c-Myc Here, I have investigated whether a heretofore underappreciated class of transcription factors, known as nuclear receptors, can function similarly to conventional pluripotency transcription factors to reprogram mouse fibroblasts into iPSCs I have identified two nuclear receptors, Nrli2 and Nr5a2, that can enhance the efficiency of iPSC generation by about 3- to 4-fold, respectively Saliently, Nr5a2 can fully replace the need for exogenous Oct4 to generate mouse iPSCs, making it the first known factor capable of “replacing” Oct4 in iPSC reprogramming Its close family member Nr5a1 functions similarly in substituting for Oct4 Piqued by how Nr5a2 can replace the singularly important Oct4 in iPSC generation, I have furthermore found that Nr5a2 is endogenously required for iPSC generation—bereft of it, few iPSCs form Moreover, in reprogramming fibroblasts, Nr5a2 directly binds the Nanog enhancer and upregulates expression of the dominant pluripotency factor Nanog In brief, my study illuminates an unexpected role for nuclear receptors in iPSC generation, identifies Nr5a2 as the first factor that viii overexpression of an ESC-important factor within EpiSCs, which totally lacks that factor to begin with187, could account for such a reprogramming feat Nonetheless, it is not surprising that the Nr5a factors are able to reprogram EpiSCs which are derived from the post-implantation stage embryo and hence their expected greater malleability to be converted (as compared to MEFs, which are typically derived from E13.5 mouse embryos, and can also be reprogrammed by Nr5a2) to iPSCs Regardless, this discovery ultimately corroborates the reprogramming capabilities of the Nr5a factors in the reprogramming of mouse cells as shown by the study herein, albeit of different developmental time point 4.6 Species-specific actions of Nr5a2 in reprogramming As the canonical reprogramming code of Oct4, Sox2, Klf4 and c-Myc can be employed to generate both mouse and human iPSCs48,49, it is of course significant to investigate if Nr5a2 also exhibits reprogramming capabilities in human cells However, what was demonstrated in murine reprogramming for Nr5a2 could not be recapitulated in human reprogramming (Figure 41) Not only was there no enhancement in the reprogramming of human fibroblasts, exogenous NR5A2 was also unable to replace exogenous OCT4 in the generation of human iPSCs (Figure 41) These results show that there is in fact some species-specific reprogramming factors, unlike what was previously thought as evidenced by the canonical reprogramming quartet However, this result is not surprising as unlike mouse ESCs, human ESCs express very minimal levels of NR5A2189 and hence introducing NR5A2, a nonhighly expressed human ESC transcription factor may not necessarily promote the 105 reversion of human somatic cells to a human ESC state Notably, human ESCs, similar to mouse EpiSCs, are also regarded to be in a primed state of pluripotency as they share many defining traits including their flattened morphology as well as their growth factor requirements in culture Incidentally, as mentioned above, EpiSCs, which are murine equivalents to human ESCs also not express Nr5a2 187 and these Nr5a2-less EpiSCs must require the exogenous introduction of this transcription factor to create a mouse ESC-like state within themselves during reprogramming187 Interestingly, in an expression profiling study of nuclear receptors performed in both mouse and human ESCs, the authors claimed species-specific functions of Nr5a2, Esrrb and Dax1, nuclear receptors that were implicated in mouse ESC biology189 These nuclear receptors that are highly expressed in mouse ESCs have different expression patterns in human ESCs Moreover, the authors also show that while Nr5a2 tends to be reduced upon differentiation in mouse ESCs, the opposite is true in human ESCs, in which its expression tends to increase upon differentiation 189 Nonetheless, it is interesting to speculate that human ESCs might contain other Nr5a2-like factors (not necessarily Nr5a2) that orchestrate cognate mechanisms as shown in mouse ESCs Taken together, it is interesting to report herein a nuclear receptor that might have a species-specific function, in which it enhances and replaces Oct4 in murine reprogramming but not exhibit the same characteristics in the context of human reprogramming 4.7 Mouse ESC-like human ESCs with Nr5a2 106 On this note, it is tempting to speculate if the overexpression of Nr5a2 might induce a mouse ESC-like state in human ESCs since this nuclear receptor can reprogram primed EpiSCs to an earlier pluripotent derivative187 Already, several groups have demonstrated the generation of mouse ESC-like human ESCs, which can be passaged as single cells, display shiny dome-shaped morphologies very akin to mouse ESCs that self-renew in culture conditions permissive for mouse ESCs190-192 These groups introduce various canonical reprogramming factors and/or a cocktail of chemical inhibitors in the presence of LIF to achieve such a reprogrammed state in human ESCs However, it should be noted that Nr5a2 appears to display species-specific roles and what is observed in the murine system may not necessarily be similarly translated for the human system In addition, Nr5a2 is also highly expressed in endodermal-derived organs and an elevation of Nr5a2 in human ESCs may induce them towards differentiation Interestingly, a group showed that NR5A2 in combination with another nuclear receptor (RARγ), in addition with OSKM, are able to reprogram human somatic cells to iPSCs that resemble mouse ESCs with respect to their morphologies, gene expression as well as culture condition requirements178 These 6-factor-reprogrammed human cells were able to be grown in LIF in the presence of 2i Notably, when the authors removed LIF and supplemented the culture media with FGF, the iPSCs assumed a hESC-like morphology instead All in all, this study demonstrates that Nr5a2 is not only able to contribute to the reprogramming of mouse somatic cells and EpiSCs to a mouse ESC-like state but also human somatic cells as well, hence these reflect the reproducibility of Nr5a2 in naïve state reprogramming 107 4.8 Possible role of Nr5a2 in transdifferentiation The potency of Nr5a2 in mediating universal reprogramming of both mouse and human cells to a mouse ESC-like state is indeed remarkable86,178,187 Incidentally, Nr5a2 is not only expressed in mouse ESCs and the early developing embryo but also in endodermal-derived adult tissues such as the liver, pancreas and intestines193-195 Besides endodermal cells, Nr5a2 is also expressed in steroidogenic tissue such as the testes and ovaries196 Hence, Nr5a2 plays a dual role in establishing pluripotency as well as inducing myriad avenues of differentiation In this respect, it is tempting to speculate the possibility of Nr5a2 in converting somatic cells such as fibroblasts into an endodermal or steroidogenic cell type It should be noted that even if it were able to cells into an endodermal derivative, there are many endodermal cell types in which Nr5a2 is expressed, and a heterogenous mixture of cells may eventually be derived Nonetheless, specialised cell culture media with relevant supplements to solely derive one endodermal cell type can be utilised Even if that is not possible, a multipotent endodermal precursor cell which Nr5a2 can create from its overexpression in fibroblast would be somewhat remarkable Fortuitously, transdifferentiation of cells to endodermal or steroidogenic tissues have yet to be demonstrated and it would therefore be interesting to investigate Nr5a2’s capabilities in orchestrating directed conversion 108 Conclusions Altogether, a screen of nuclear receptors in their potential ability to augment the generation of mouse iPSCs reveals nuclear receptors Nr1i2 and Nr5a2 as enhancers of reprogramming Besides being able to enhance the number of iPSC colonies generated, Nr5a2 is also able to enhance the kinetics of reprogramming Strikingly, Nr5a2 can replace exogenous Oct4 in the reprogramming of MEFs This finding is novel as no transcription factor, let alone factor have been previously shown to be able to substitute for exogenous Oct4 in the reprogramming process Interestingly, besides commonly known to work through Oct4, Nr5a2 is herein shown to also work in part through Nanog to mediate the successful reprogramming of murine somatic cells In summary, the nuclear receptor Nr5a2 is indeed a potent reprogramming factor that can partake in the reprogramming process to generate iPSCs, and more importantly recreate a state of pluripotent naivety that greatly resembles mouse ESCs 109 References 10 11 12 13 14 15 16 17 18 19 20 21 Johnson, K.M., Mitsouras, K & Carey, M Eukaryotic transcription: the core of eukaryotic gene activation Curr Biol 11, R510-3 (2001) Levine, M & Tjian, R Transcription regulation and animal diversity Nature 424, 147-51 (2003) Lander, E.S et al Initial sequencing and analysis of the human genome Nature 409, 860-921 (2001) Bernstein, B.E et al A bivalent chromatin structure marks key developmental genes in embryonic stem cells Cell 125, 315-26 (2006) Peters, A.H et al Histone H3 lysine methylation is an epigenetic imprint of facultative heterochromatin Nat Genet 30, 77-80 (2002) Boyer, L.A et al Polycomb complexes repress developmental regulators in murine embryonic stem cells Nature 441, 349-53 (2006) Cao, R et al Role of histone H3 lysine 27 methylation in Polycomb-group silencing Science 298, 1039-43 (2002) Grau, D.J et al Compaction of chromatin by diverse Polycomb group proteins requires localized regions of high charge Genes Dev 25, 2210-21 (2011) Francis, N.J., Kingston, R.E & Woodcock, C.L Chromatin compaction by a polycomb group protein complex Science 306, 1574-7 (2004) Schuettengruber, B., Martinez, A.M., Iovino, N & Cavalli, G Trithorax group proteins: switching genes on and keeping them active Nat Rev Mol Cell Biol 12, 799-814 (2011) Brook, F.A & Gardner, R.L The origin and efficient derivation of embryonic stem cells in the mouse Proc Natl Acad Sci U S A 94, 5709-12 (1997) Pesce, M & Scholer, H.R Oct-4: gatekeeper in the beginnings of mammalian development Stem Cells 19, 271-8 (2001) Nichols, J et al Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4 Cell 95, 379-91 (1998) Niwa, H., Miyazaki, J & Smith, A.G Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells Nat Genet 24, 372-6 (2000) Episkopou, V SOX2 functions in adult neural stem cells Trends Neurosci 28, 219-21 (2005) Avilion, A.A et al Multipotent cell lineages in early mouse development depend on SOX2 function Genes Dev 17, 126-40 (2003) Masui, S et al Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells Nat Cell Biol 9, 625-35 (2007) Kopp, J.L., Ormsbee, B.D., Desler, M & Rizzino, A Small increases in the level of Sox2 trigger the differentiation of mouse embryonic stem cells Stem Cells 26, 903-911 (2008) Nishimoto, M., Fukushima, A., Okuda, A & Muramatsu, M The gene for the embryonic stem cell coactivator UTF1 carries a regulatory element which selectively interacts with a complex composed of Oct-3/4 and Sox-2 Mol Cell Biol 19, 5453-65 (1999) Rodda, D.J et al Transcriptional regulation of nanog by OCT4 and SOX2 J Biol Chem 280, 24731-7 (2005) Kuroda, T et al Octamer and Sox elements are required for transcriptional cis regulation of Nanog gene expression Mol Cell Biol 25, 2475-85 (2005) 110 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 Ambrosetti, D.C., Basilico, C & Dailey, L Synergistic activation of the fibroblast growth factor enhancer by Sox2 and Oct-3 depends on proteinprotein interactions facilitated by a specific spatial arrangement of factor binding sites Mol Cell Biol 17, 6321-9 (1997) Yamaguchi, S., Kimura, H., Tada, M., Nakatsuji, N & Tada, T Nanog expression in mouse germ cell development Gene Expr Patterns 5, 639-46 (2005) Chambers, I et al Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells Cell 113, 643-55 (2003) Mitsui, K et al The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells Cell 113, 631-42 (2003) Wang, J., Levasseur, D.N & Orkin, S.H Requirement of Nanog dimerization for stem cell self-renewal and pluripotency Proc Natl Acad Sci U S A 105, 6326-31 (2008) Chambers, I et al Nanog safeguards pluripotency and mediates germline development Nature 450, 1230-4 (2007) Silva, J et al Nanog is the gateway to the pluripotent ground state Cell 138, 722-37 (2009) Lim, L.S et al Zic3 is required for maintenance of pluripotency in embryonic stem cells Mol Biol Cell 18, 1348-58 (2007) Dejosez, M et al Ronin is essential for embryogenesis and the pluripotency of mouse embryonic stem cells Cell 133, 1162-74 (2008) Zhang, J et al Sall4 modulates embryonic stem cell pluripotency and early embryonic development by the transcriptional regulation of Pou5f1 Nat Cell Biol 8, 1114-23 (2006) Ivanova, N et al Dissecting self-renewal in stem cells with RNA interference Nature 442, 533-8 (2006) Evans, M.J & Kaufman, M.H Establishment in culture of pluripotential cells from mouse embryos Nature 292, 154-6 (1981) Martin, G.R Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells Proc Natl Acad Sci U S A 78, 7634-8 (1981) Bongso, A., Fong, C.Y., Ng, S.C & Ratnam, S Isolation and culture of inner cell mass cells from human blastocysts Hum Reprod 9, 2110-7 (1994) Thomson, J.A et al Embryonic stem cell lines derived from human blastocysts Science 282, 1145-7 (1998) Hall, B.K In search of evolutionary developmental mechanisms: the 30-year gap between 1944 and 1974 J Exp Zool B Mol Dev Evol 302, 5-18 (2004) Yu, J., Vodyanik, M.A., He, P., Slukvin, II & Thomson, J.A Human embryonic stem cells reprogram myeloid precursors following cell-cell fusion Stem Cells 24, 168-76 (2006) Cowan, C.A., Atienza, J., Melton, D.A & Eggan, K Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells Science 309, 1369-73 (2005) Wilmut, I., Schnieke, A.E., McWhir, J., Kind, A.J & Campbell, K.H Viable offspring derived from fetal and adult mammalian cells Nature 385, 810-3 (1997) Lee, J.B & Park, C Molecular genetics: verification that Snuppy is a clone Nature 440, E2-3 (2006) 111 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 Matsui, Y., Zsebo, K & Hogan, B.L Derivation of pluripotential embryonic stem cells from murine primordial germ cells in culture Cell 70, 841-7 (1992) Kanatsu-Shinohara, M et al Generation of pluripotent stem cells from neonatal mouse testis Cell 119, 1001-12 (2004) Guan, K et al Pluripotency of spermatogonial stem cells from adult mouse testis Nature 440, 1199-203 (2006) Allen, N.D., Barton, S.C., Hilton, K., Norris, M.L & Surani, M.A A functional analysis of imprinting in parthenogenetic embryonic stem cells Development 120, 1473-82 (1994) Cibelli, J.B et al Parthenogenetic stem cells in nonhuman primates Science 295, 819 (2002) Hernandez, L., Kozlov, S., Piras, G & Stewart, C.L Paternal and maternal genomes confer opposite effects on proliferation, cell-cycle length, senescence, and tumor formation Proc Natl Acad Sci U S A 100, 13344-9 (2003) Takahashi, K & Yamanaka, S Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors Cell 126, 663-76 (2006) Takahashi, K et al Induction of pluripotent stem cells from adult human fibroblasts by defined factors Cell 131, 861-72 (2007) Park, I.H et al Reprogramming of human somatic cells to pluripotency with defined factors Nature 451, 141-6 (2008) Yu, J et al Induced pluripotent stem cell lines derived from human somatic cells Science 318, 1917-20 (2007) Feng, B., Ng, J.H., Heng, J.C & Ng, H.H Molecules that promote or enhance reprogramming of somatic cells to induced pluripotent stem cells Cell Stem Cell 4, 301-12 (2009) Boyer, L.A et al Core transcriptional regulatory circuitry in human embryonic stem cells Cell 122, 947-56 (2005) Loh, Y.H et al The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells Nat Genet 38, 431-40 (2006) Nakagawa, M et al Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts Nat Biotechnol 26, 101-6 (2008) Eminli, S., Utikal, J., Arnold, K., Jaenisch, R & Hochedlinger, K Reprogramming of neural progenitor cells into induced pluripotent stem cells in the absence of exogenous Sox2 expression Stem Cells 26, 2467-74 (2008) Ichida, J.K et al A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog Cell Stem Cell 5, 491-503 (2009) Jiang, J et al A core Klf circuitry regulates self-renewal of embryonic stem cells Nat Cell Biol 10, 353-60 (2008) Parisi, S et al Klf5 is involved in self-renewal of mouse embryonic stem cells J Cell Sci 121, 2629-34 (2008) Ema, M et al Kruppel-like factor is essential for blastocyst development and the normal self-renewal of mouse ESCs Cell Stem Cell 3, 555-67 (2008) Hall, J et al Oct4 and LIF/Stat3 additively induce Kruppel factors to sustain embryonic stem cell self-renewal Cell Stem Cell 5, 597-609 (2009) Lyssiotis, C.A et al Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4 Proc Natl Acad Sci U S A 106, 8912-7 (2009) 112 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 Rowland, B.D., Bernards, R & Peeper, D.S The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene Nat Cell Biol 7, 1074-82 (2005) Varlakhanova, N.V et al myc maintains embryonic stem cell pluripotency and self-renewal Differentiation 80, 9-19 (2010) Knoepfler, P.S et al Myc influences global chromatin structure Embo J 25, 2723-34 (2006) Wernig, M., Meissner, A., Cassady, J.P & Jaenisch, R c-Myc Is Dispensable for Direct Reprogramming of Mouse Fibroblasts Cell Stem Cell 2, 10-12 (2008) Sridharan, R et al Role of the murine reprogramming factors in the induction of pluripotency Cell 136, 364-77 (2009) Nakagawa, M., Takizawa, N., Narita, M., Ichisaka, T & Yamanaka, S Promotion of direct reprogramming by transformation-deficient Myc Proc Natl Acad Sci U S A 107, 14152-7 (2010) Zhao, T., Zhang, Z.N., Rong, Z & Xu, Y Immunogenicity of induced pluripotent stem cells Nature 474, 212-5 (2011) Hanna, J et al Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin Science 318, 1920-3 (2007) Zhang, J et al A human iPSC model of Hutchinson Gilford Progeria reveals vascular smooth muscle and mesenchymal stem cell defects Cell Stem Cell 8, 31-45 (2011) Liu, G.H et al Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome Nature 472, 221-5 (2011) Carvajal-Vergara, X et al Patient-specific induced pluripotent stem-cellderived models of LEOPARD syndrome Nature 465, 808-12 (2010) Dimos, J.T et al Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons Science 321, 1218-21 (2008) Feldman, N et al G9a-mediated irreversible epigenetic inactivation of Oct3/4 during early embryogenesis Nat Cell Biol 8, 188-94 (2006) Silva, J et al Promotion of reprogramming to ground state pluripotency by signal inhibition PLoS Biol 6, e253 (2008) Mikkelsen, T.S et al Dissecting direct reprogramming through integrative genomic analysis Nature 454, 49-55 (2008) Li, R et al A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts Cell Stem Cell 7, 51-63 (2010) Samavarchi-Tehrani, P et al Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming Cell Stem Cell 7, 64-77 (2010) Stadtfeld, M., Maherali, N., Breault, D.T & Hochedlinger, K Defining Molecular Cornerstones during Fibroblast to iPS Cell Reprogramming in Mouse Cell Stem Cell 2, 230-240 (2008) Okita, K., Ichisaka, T & Yamanaka, S Generation of germline-competent induced pluripotent stem cells Nature 448, 313-7 (2007) Wernig, M et al In vitro reprogramming of fibroblasts into a pluripotent EScell-like state Nature 448, 318-24 (2007) Meissner, A., Wernig, M & Jaenisch, R Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells Nat Biotechnol 25, 1177-81 (2007) 113 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 Blelloch, R., Venere, M., Yen, J & Ramalho-Santos, M Generation of Induced Pluripotent Stem Cells in the Absence of Drug Selection Cell Stem Cell 1, 245-247 (2007) Feng, B et al Reprogramming of fibroblasts into induced pluripotent stem cells with orphan nuclear receptor Esrrb Nat Cell Biol 11, 197-203 (2009) Heng, J.C et al The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells Cell Stem Cell 6, 167-74 (2010) Kim, J.B et al Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors Nature 454, 646-50 (2008) Boland, M.J et al Adult mice generated from induced pluripotent stem cells Nature 461, 91-4 (2009) Zhao, X.Y et al iPS cells produce viable mice through tetraploid complementation Nature 461, 86-90 (2009) Kang, L., Wang, J., Zhang, Y., Kou, Z & Gao, S iPS cells can support fullterm development of tetraploid blastocyst-complemented embryos Cell Stem Cell 5, 135-8 (2009) Hanna, J et al Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency Cell 133, 250-64 (2008) Aoi, T et al Generation of pluripotent stem cells from adult mouse liver and stomach cells Science 321, 699-702 (2008) Aasen, T et al Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes Nat Biotechnol 26, 1276-84 (2008) Shi, Y et al A combined chemical and genetic approach for the generation of induced pluripotent stem cells Cell Stem Cell 2, 525-8 (2008) Kim, J.B et al Direct reprogramming of human neural stem cells by OCT4 Nature 461, 649-3 (2009) Kim, J.B et al Oct4-induced pluripotency in adult neural stem cells Cell 136, 411-9 (2009) Hanna, J et al Metastable pluripotent states in NOD-mouse-derived ESCs Cell Stem Cell 4, 513-24 (2009) Park, I.H., Lerou, P.H., Zhao, R., Huo, H & Daley, G.Q Generation of human-induced pluripotent stem cells Nat Protoc 3, 1180-6 (2008) Li, W et al Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors Cell Stem Cell 4, 16-9 (2009) Liao, J et al Generation of induced pluripotent stem cell lines from adult rat cells Cell Stem Cell 4, 11-5 (2009) Ezashi, T et al Derivation of induced pluripotent stem cells from pig somatic cells Proc Natl Acad Sci U S A 106, 10993-8 (2009) Wu, Z et al Generation of pig induced pluripotent stem cells with a druginducible system J Mol Cell Biol 1, 46-54 (2009) Esteban, M.A et al Generation of induced pluripotent stem cell lines from Tibetan miniature pig J Biol Chem 284, 17634-40 (2009) Liu, H et al Generation of Induced Pluripotent Stem Cells from Adult Rhesus Monkey Fibroblasts Cell Stem Cell 3, 587-590 (2008) Honda, A et al Generation of induced pluripotent stem cells in rabbits: potential experimental models for human regenerative medicine J Biol Chem 285, 31362-9 (2010) 114 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 Brambrink, T et al Sequential Expression of Pluripotency Markers during Direct Reprogramming of Mouse Somatic Cells Cell Stem Cell 2, 151-159 (2008) Stadtfeld, M., Nagaya, M., Utikal, J., Weir, G & Hochedlinger, K Induced pluripotent stem cells generated without viral integration Science 322, 945-9 (2008) Okita, K., Nakagawa, M., Hyenjong, H., Ichisaka, T & Yamanaka, S Generation of mouse induced pluripotent stem cells without viral vectors Science 322, 949-53 (2008) Gonzalez, F et al Generation of mouse-induced pluripotent stem cells by transient expression of a single nonviral polycistronic vector Proc Natl Acad Sci U S A 106, 8918-22 (2009) Woltjen, K et al piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells Nature 458, 766-70 (2009) Kaji, K et al Virus-free induction of pluripotency and subsequent excision of reprogramming factors Nature 458, 771-5 (2009) Yusa, K., Rad, R., Takeda, J & Bradley, A Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon Nat Methods 6, 363-9 (2009) Jia, F et al A nonviral minicircle vector for deriving human iPS cells Nat Methods 7, 197-9 (2010) Kim, D et al Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins Cell Stem Cell 4, 472-6 (2009) Zhou, H et al Generation of induced pluripotent stem cells using recombinant proteins Cell Stem Cell 4, 381-4 (2009) Warren, L et al Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA Cell Stem Cell 7, 618-30 (2010) Diebold, S.S., Kaisho, T., Hemmi, H., Akira, S & Reis e Sousa, C Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA Science 303, 1529-31 (2004) Hornung, V et al 5'-Triphosphate RNA is the ligand for RIG-I Science 314, 994-7 (2006) Burdon, T., Stracey, C., Chambers, I., Nichols, J & Smith, A Suppression of SHP-2 and ERK signalling promotes self-renewal of mouse embryonic stem cells Dev Biol 210, 30-43 (1999) Cheng, A.M et al Mammalian Grb2 regulates multiple steps in embryonic development and malignant transformation Cell 95, 793-803 (1998) Miyamoto, Y., Yamauchi, J., Mizuno, N & Itoh, H The adaptor protein Nck1 mediates endothelin A receptor-regulated cell migration through the Cdc42dependent c-Jun N-terminal kinase pathway J Biol Chem 279, 34336-42 (2004) Chang, D.W., Claassen, G.F., Hann, S.R & Cole, M.D The c-Myc transactivation domain is a direct modulator of apoptotic versus proliferative signals Mol Cell Biol 20, 4309-19 (2000) Bromberg, J.F et al Stat3 as an oncogene Cell 98, 295-303 (1999) Sadot, E et al Regulation of S33/S37 phosphorylated beta-catenin in normal and transformed cells J Cell Sci 115, 2771-80 (2002) 115 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 Tokuzawa, Y et al Fbx15 Is a Novel Target of Oct3/4 but Is Dispensable for Embryonic Stem Cell Self-Renewal and Mouse Development Mol Cell Biol 23, 2699-2708 (2003) Newman, M.A., Thomson, J.M & Hammond, S.M Lin-28 interaction with the Let-7 precursor loop mediates regulated microRNA processing Rna 14, 1539-49 (2008) Viswanathan, S.R., Daley, G.Q & Gregory, R.I Selective blockade of microRNA processing by Lin28 Science 320, 97-100 (2008) Yu, F et al let-7 regulates self renewal and tumorigenicity of breast cancer cells Cell 131, 1109-23 (2007) West, J.A et al A role for Lin28 in primordial germ-cell development and germ-cell malignancy Nature 460, 909-13 (2009) Hanna, J et al Direct cell reprogramming is a stochastic process amenable to acceleration Nature 462, 595-601 (2009) Zhao, Y et al Two supporting factors greatly improve the efficiency of human iPSC generation Cell Stem Cell 3, 475-9 (2008) Kooistra, S.M et al Undifferentiated embryonic cell transcription factor regulates ESC chromatin organization and gene expression Stem Cells 28, 1703-14 (2010) Kawamura, T et al Linking the p53 tumour suppressor pathway to somatic cell reprogramming Nature 460, 1140-4 (2009) Marion, R.M et al A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity Nature 460, 1149-53 (2009) Utikal, J et al Immortalization eliminates a roadblock during cellular reprogramming into iPS cells Nature 460, 1145-8 (2009) Li, H et al The Ink4/Arf locus is a barrier for iPS cell reprogramming Nature 460, 1136-9 (2009) Hong, H et al Suppression of induced pluripotent stem cell generation by the p53-p21 pathway Nature 460, 1132-5 (2009) Mali, P et al Improved efficiency and pace of generating induced pluripotent stem cells from human adult and fetal fibroblasts Stem Cells 26, 1998-2005 (2008) Hahn, W.C et al Creation of human tumour cells with defined genetic elements Nature 400, 464-8 (1999) Ahuja, D., Saenz-Robles, M.T & Pipas, J.M SV40 large T antigen targets multiple cellular pathways to elicit cellular transformation Oncogene 24, 7729-45 (2005) Maherali, N et al Directly Reprogrammed Fibroblasts Show Global Epigenetic Remodeling and Widespread Tissue Contribution Cell Stem Cell 1, 55-70 (2007) Zhao, R & Daley, G.Q From fibroblasts to iPS cells: induced pluripotency by defined factors J Cell Biochem 105, 949-55 (2008) Marion, R.M et al Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells Cell Stem Cell 4, 141-54 (2009) Jauch, R et al Conversion of Sox17 into a pluripotency reprogramming factor by reengineering its association with Oct4 on DNA Stem Cells 29, 940-51 (2011) Maekawa, M et al Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1 Nature 474, 225-9 (2011) 116 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 Bartel, D.P MicroRNAs: target recognition and regulatory functions Cell 136, 215-33 (2009) Li, Z., Yang, C.S., Nakashima, K & Rana, T.M Small RNA-mediated regulation of iPS cell generation EMBO J 30, 823-34 (2011) Lin, S.L et al Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state Rna 14, 2115-24 (2008) Marson, A et al Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells Cell 134, 521-33 (2008) Houbaviy, H.B., Murray, M.F & Sharp, P.A Embryonic stem cell-specific MicroRNAs Dev Cell 5, 351-8 (2003) Liao, B et al MicroRNA cluster 302-367 enhances somatic cell reprogramming by accelerating a mesenchymal-to-epithelial transition J Biol Chem 286, 17359-64 (2011) Anokye-Danso, F et al Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency Cell Stem Cell 8, 376-88 (2011) Miyoshi, N et al Reprogramming of mouse and human cells to pluripotency using mature microRNAs Cell Stem Cell 8, 633-8 (2011) Pfaff, N et al miRNA screening reveals a new miRNA family stimulating iPS cell generation via regulation of Meox2 EMBO Rep 12, 1153-9 (2011) Ponting, C.P., Oliver, P.L & Reik, W Evolution and functions of long noncoding RNAs Cell 136, 629-41 (2009) Guttman, M et al Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals Nature 458, 223-7 (2009) Loewer, S et al Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells Nat Genet 42, 11137 (2010) Krizhanovsky, V & Lowe, S.W Stem cells: The promises and perils of p53 Nature 460, 1085-6 (2009) Mullen, E.M., Gu, P & Cooney, A.J Nuclear Receptors in Regulation of Mouse ES Cell Pluripotency and Differentiation PPAR Res 2007, 61563 (2007) Olefsky, J.M Nuclear receptor minireview series J Biol Chem 276, 36863-4 (2001) A unified nomenclature system for the nuclear receptor superfamily Cell 97, 161-3 (1999) Mukherjee, R., Jow, L., Croston, G.E & Paterniti, J.R., Jr Identification, characterization, and tissue distribution of human peroxisome proliferatoractivated receptor (PPAR) isoforms PPARgamma2 versus PPARgamma1 and activation with retinoid X receptor agonists and antagonists J Biol Chem 272, 8071-6 (1997) Lan, Z.J., Gu, P., Xu, X & Cooney, A.J Expression of the orphan nuclear receptor, germ cell nuclear factor, in mouse gonads and preimplantation embryos Biol Reprod 68, 282-9 (2003) Allenby, G et al Retinoic acid receptors and retinoid X receptors: interactions with endogenous retinoic acids Proc Natl Acad Sci U S A 90, 30-4 (1993) Gu, P., Le Menuet, D., Chung, A.C & Cooney, A.J Differential recruitment of methylated CpG binding domains by the orphan receptor GCNF initiates the repression and silencing of Oct4 expression Mol Cell Biol 26, 9471-83 (2006) 117 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 Gu, P et al Orphan nuclear receptor LRH-1 is required to maintain Oct4 expression at the epiblast stage of embryonic development Mol Cell Biol 25, 3492-505 (2005) Wang, J et al A protein interaction network for pluripotency of embryonic stem cells Nature 444, 364-8 (2006) Clipsham, R., Niakan, K & McCabe, E.R Nr0b1 and its network partners are expressed early in murine embryos prior to steroidogenic axis organogenesis Gene Expr Patterns 4, 3-14 (2004) Yang, F.M et al Liver receptor homolog-1 localization in the nuclear body is regulated by sumoylation and cAMP signaling in rat granulosa cells Febs J 276, 425-36 (2009) Morita, S., Kojima, T & Kitamura, T Plat-E: an efficient and stable system for transient packaging of retroviruses Gene Ther 7, 1063-6 (2000) Tiscornia, G., Singer, O & Verma, I.M Production and purification of lentiviral vectors Nat Protoc 1, 241-5 (2006) Wang, W & Lufkin, T The murine Otp homeobox gene plays an essential role in the specification of neuronal cell lineages in the developing hypothalamus Dev Biol 227, 432-49 (2000) Tusher, V.G., Tibshirani, R & Chu, G Significance analysis of microarrays applied to the ionizing radiation response Proc Natl Acad Sci U S A 98, 511621 (2001) Pasque, V., Miyamoto, K & Gurdon, J.B Efficiencies and mechanisms of nuclear reprogramming Cold Spring Harb Symp Quant Biol 75, 189-200 (2010) Yeom, Y.I et al Germline regulatory element of Oct-4 specific for the totipotent cycle of embryonal cells Development 122, 881-94 (1996) Chen, X et al Integration of external signaling pathways with the core transcriptional network in embryonic stem cells Cell 133, 1106-17 (2008) Han, J et al Tbx3 improves the germ-line competency of induced pluripotent stem cells Nature 463, 1096-100 (2010) Wang, W et al Rapid and efficient reprogramming of somatic cells to induced pluripotent stem cells by retinoic acid receptor gamma and liver receptor homolog Proc Natl Acad Sci U S A 108, 18283-8 (2011) Ihunnah, C.A., Jiang, M & Xie, W Nuclear receptor PXR, transcriptional circuits and metabolic relevance Biochim Biophys Acta 1812, 956-63 (2011) Sadovsky, Y et al Mice deficient in the orphan receptor steroidogenic factor lack adrenal glands and gonads but express P450 side-chain-cleavage enzyme in the placenta and have normal embryonic serum levels of corticosteroids Proc Natl Acad Sci U S A 92, 10939-43 (1995) Maherali, N & Hochedlinger, K Tgfbeta signal inhibition cooperates in the induction of iPSCs and replaces Sox2 and cMyc Curr Biol 19, 1718-23 (2009) Li, W et al Generation of human-induced pluripotent stem cells in the absence of exogenous Sox2 Stem Cells 27, 2992-3000 (2009) Yuan, X et al Combined chemical treatment enables Oct4-induced reprogramming from mouse embryonic fibroblasts Stem Cells 29, 549-53 (2011) Zhu, S et al Reprogramming of human primary somatic cells by OCT4 and chemical compounds Cell Stem Cell 7, 651-5 (2010) 118 185 186 187 188 189 190 191 192 193 194 195 196 Pare, J.F et al The fetoprotein transcription factor (FTF) gene is essential to embryogenesis and cholesterol homeostasis and is regulated by a DR4 element J Biol Chem 279, 21206-16 (2004) Labelle-Dumais, C., Jacob-Wagner, M., Pare, J.F., Belanger, L & Dufort, D Nuclear receptor NR5A2 is required for proper primitive streak morphogenesis Dev Dyn 235, 3359-69 (2006) Guo, G & Smith, A A genome-wide screen in EpiSCs identifies Nr5a nuclear receptors as potent inducers of ground state pluripotency Development 137, 3185-92 (2010) Nichols, J & Smith, A Naive and primed pluripotent states Cell Stem Cell 4, 487-92 (2009) Xie, C.Q et al Expression profiling of nuclear receptors in human and mouse embryonic stem cells Mol Endocrinol 23, 724-33 (2009) Hanna, J et al Human embryonic stem cells with biological and epigenetic characteristics similar to those of mouse ESCs Proc Natl Acad Sci U S A 107, 9222-7 (2010) Xu, Y et al Revealing a core signaling regulatory mechanism for pluripotent stem cell survival and self-renewal by small molecules Proc Natl Acad Sci U S A 107, 8129-34 (2010) Buecker, C et al A Murine ESC-like State Facilitates Transgenesis and Homologous Recombination in Human Pluripotent Stem Cells Cell Stem Cell 6, 535-546 (2010) Annicotte, J.S et al Pancreatic-duodenal homeobox regulates expression of liver receptor homolog during pancreas development Mol Cell Biol 23, 6713-24 (2003) Nitta, M., Ku, S., Brown, C., Okamoto, A.Y & Shan, B CPF: an orphan nuclear receptor that regulates liver-specific expression of the human cholesterol 7alpha-hydroxylase gene Proc Natl Acad Sci U S A 96, 6660-5 (1999) Rausa, F.M., Galarneau, L., Belanger, L & Costa, R.H The nuclear receptor fetoprotein transcription factor is coexpressed with its target gene HNF-3beta in the developing murine liver, intestine and pancreas Mech Dev 89, 185-8 (1999) Sirianni, R et al Liver receptor homologue-1 is expressed in human steroidogenic tissues and activates transcription of genes encoding steroidogenic enzymes J Endocrinol 174, R13-7 (2002) 119 .. .SCREEN OF NUCLEAR RECEPTORS FOR THE ENHANCED AND ALTERNATIVE GENERATION OF INDUCED PLURIPOTENT STEM CELLS DOMINIC HENG JIAN CHIEN (B.Sc (Hons), NTU) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR... akin to pluripotent embryonic stem cells (ESCs) and pluripotent cells of the early embryo, thus gaining their characteristic developmental potential and other characteristics diagnostic of pluripotent. .. cells via the process of pluripotential reprogramming in which the lineage restriction of differentiated cells is undone The resultant cells, known colloquially as induced pluripotent stem cells

Ngày đăng: 09/09/2015, 18:56

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan