Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Apr 26, 2015; 7(3): 541-546
Published online Apr 26, 2015. doi: 10.4252/wjsc.v7.i3.541
Repressors of reprogramming
Melissa Popowski, the Rockefeller University, New York, NY 10065, United States
Haley Tucker, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78701, United States
Author contributions: Popowski M and Tucker H conceived and wrote the review.
Conflict-of-interest: The authors declare no conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Haley Tucker, PhD, Institute for Cellular and Molecular Biology, University of Texas at Austin, 1710 Red River, Austin, TX 78701, United States. haleytucker@austin.utexas.edu
Telephone: +1-512-4757705 Fax: +1-512-4757707
Received: July 31, 2014
Peer-review started: August 1, 2014
First decision: September 4, 2014
Revised: January 10, 2015
Accepted: January 18, 2015
Article in press: January 20, 2015
Published online: April 26, 2015

Abstract

Induced pluripotent stem cells (iPSCs) have been the focal point of ever increasing interest and scrutiny as they hold the promise of personalized regenerative medicine. However, creation of iPSCs is an inefficient process that requires forced expression of potentially oncogenic proteins. In order to unlock the full potential of iPSCs, both for basic and clinical research, we must broaden our search for more reliable ways of inducing pluripotency in somatic cells. This review surveys an area of reprogramming that does not receive as much focus, barriers to reprogramming, in the hope of stimulating new ideas and approaches towards developing safer and more efficient methods of reprogramming. Better methods of iPSC creation will allow for more reliable disease modeling, better basic research into the pluripotent state and safer iPSCs that can be used in a clinical setting.

Key Words: Reprogramming, Induced pluripotency, Stem cells

Core tip: This review addresses an underappreciated aspect of cellular reprogramming, repressors of reprograming. We review current literature focusing on inhibitors that modify cellular reprogramming.



INTRODUCTION

The interest in cellular reprogramming spans basic science to clinical studies with the aim to understand the pluripotent state as well as to harness its potential to generate patient-specific therapies. These goals, while lofty, seem increasingly within reach. The first evidence of somatic cellular reprogramming was provided by the experiments performed by Gurdon et al[1,2] which employed somatic cell nuclear transfer (SCNT) in Xenopus laevis. SCNT was later shown to be effective in mammals with the cloning of “Dolly” the sheep[3,4] and has recently been shown to work in humans[5,6]. SCNT requires enucleated oocytes to mediate virtually complete reprogramming of differentiated nuclei. Induced pluripotent stem cells (iPSC) were first described by Takahashi et al[7,8] by overexpressing Oct4, Sox2, c-Myc and Klf4-transcription factors shown to be essential for embryonic stem cell pluripotency. The discovery that forced expression of defined factors in differentiated cells could reprogram them to a stem-cell like state has had an enormous impact on the field of stem cell research. Advances in the creation of disease-specific stem cells are allowing us to gain previously unobtainable insights into a wide variety of diseases. Understanding the mechanics of cellular reprogramming will allow a greater understanding of developmental biology but will also translate into safe, reliable patient-specific cellular therapies.

Reprogramming is often thought of as a forward-moving process that relies on exposing the differentiated nucleus to factors that induce de-differentiation. Many groups have made strides in improving the methods of enforcing the expression of reprogramming factors[9], increasing their efficiency[10-12] and employing alternative reprogramming factors[13-15]. Problematically, iPSC and to a lesser extent even SCNT-derived stem cells have been shown to be incompletely reprogrammed, leading to possible differentiation bias or unregulated growth during later differentiation[16]. Incomplete reprogramming may also lead to unknown biases when studying disease-specific stem cells, potentially confounding results. Efficient, safe, and completely reprogrammed cells are a necessary step towards moving iPSC into the clinic. Toward this end, far less attention has been focused on eliminating endogenous factors (or their activities) which enhance cellular differentiation. Inhibition of these “negative” factors, rather than or in combination with enforcement of positive-acting factors, could lead to safer, more efficient and more completely reprogrammed iPSC. Below we summarize a number of such targets, including, as best understood, their mechanisms and their pros and cons for clinical implementation.

TRANSCRIPTION FACTORS

After the first description of iPSC, focus was turned towards finding modifiers of reprogramming. Repression of p53[17], as well as its primary target gene p21, or addition of mutant p53[18] markedly facilitates and expedites reprogramming of mouse and human cells[19-23]. These studies proposed mechanisms by which p53 inhibits iPSCs generation, including induction of cell-cycle arrest, apoptosis and senescence. By virtue of its apoptotic induction following cellular DNA damage, p53 is a master regulator of genomic integrity and serves as a major tumor suppressor[24-26]. p53, either directly or via pathway members, undergoes loss of function mutations in nearly every cancer. Often the cellular programs that are co-opted during the development of cancer are, in fact, reactivated features of stem cells[27,28]. Reprograming differentiated cells to pluripotent cells involves massive genomic changes that are not unlike some of the genomic changes that occur during cancer development. Indeed, despite the ability of p53 repression to enhance reprogramming efficiency, p53-deficient cells can yield defective iPSCs with chromosomal aberrations, genome instability and malignant transformation[18,20,29], further highlighting the link between reprogramming and tumorigenesis. Genomic integrity is a necessity for somatic cell reprogramming in order to create viable iPSC cells as well as in maintaining cellular identity. Inhibition of p53, while an interesting target for repression because of its enhancement of reprogramming, has inherent and serious risks.

The transcription factor Bright/ARID3A regulates reprogramming through direct de-repression of key pluripotency factors Oct4, Sox2 and Nanog[30]. It has been shown to be a binding partner of Nanog[31] as well as Oct4 and Sox2[30] and is believed to function as a direct repressor of key developmental promoters. In culture, loss of Bright, either in through germline deletion, dominant negative hibition, or shRNA mediated knockdown, is able to enhance standard reprogramming, induce pluripotency related genes and partially reprogram somatic cells to an ES-like state. Further, loss of Bright can induce complete reprogramming in the presence of LIF[30]. Bright was first discovered as a B-cell specific factor[32-34] and is critical for hematopoietic stem cell formation[35]. Bright is known to interact directly with p53[36] and is regulated by p53[37]. It is possible that part of p53’s influence on reprogramming may be a function of its regulation of Bright. Loss of Bright has further been shown to inhibit senescence in culture[38], perhaps enhancing its reprogramming abilities. Bright is a tantalizing target for further analysis but there may be still as yet unknown factors which, like Bright, have multiple functions that work towards maintaining the differentiated state of the cell.

EPIGENETIC MODIFIERS

Epigenetic regulation of the genome is essential for proper gene expression and lineage commitment. As cells shift from a stem cell state into a lineage committed state, the genome goes through massive epigenetic changes from an “open” conformation to a “closed” conformation[39]. During differentiation, global acetylation marks, typically associated with euchromatin, are reduced and global methylation markers that are associated with condensed or heterochromatin, are increased[40], including H3K27[41,42] and H3K9[43,44] methylation, typically associated with inactive promoters and H3K4 methylation[45,46], which is associated with active promoter regions. Heterochromatin is tightly packed chromatin that generally inhibits gene expression. This shift in chromatin structure and epigenetic marks is a necessary part of the differentiation process, silencing of genes that are no longer expressed in that tissue type. Underlying the critical nature of these epigenetic marks, it is often incorrect expression of silenced genes that contribute to the formation of cancer[47], demonstrating their importance in gene regulation. Reprogramming can occur when “pioneer” factors such as Oct4, Sox2, and Klf4 bind to enhancers near silenced chromatin and can then direct the binding of other transcription factors to these regions[48]. Regions of the genome in fibroblasts, marked by H3K9me3, have been shown to be refractory to binding by Oct4, Sox2 and Klf4, however knockdown of H3K9 methylases increased reprogramming efficiency[49] and the binding of “pioneer” factors to these regions[50]. Thus, the structure and epigenetic marks of heterochromatin itself inhibits reprogramming by exogenous factors.

Stem cells often use bivalent domains to create “poised” promoters of important developmental genes[51]. These bivalent domains are characterized by the repressive H3K27me3 and the activating H3K4me3 marks, thus repressing expression of these genes but allowing for quick upregulation in response to developmental signals[52]. Bivalent domains are an important regulatory feature, allowing stem cells to repress the expression of key genes while keeping them “poised”. Epigenetic regulation of the genome is necessary for appropriate gene expression in both stem cells and differentiated cells[40,41] and thus erasure of established epigenetic marks is a necessary part of reprogramming. Several histone deacetylase inhibitors have been shown to modulate reprogramming in both SCNT and iPSC[53]. Valproic acid (VPA) is an extremely effective histone deacetylase (HDAC) inhibitor[54] and is commonly used to enhance reprogramming[55,56]. Other HDAC inhibitors have similarly been shown to enhance reprogramming in many different contexts[53]. The complexities of the histone code, in particular methylation of histones, leads to an unclear picture of how inhibitors of methylation effects reprogramming[57]. However, DNA demethylation agent 5’-Azacytidine has long been known to enhance reprogramming[58]. Repression of AID, a cytidine deaminase, inhibits reprogramming in interspecies heterokaryons[59] via promoter CpG demethylation[59,60]. Inhibition of both DNA and histone methylating components of the polycomb repressive complexes 1 and 2 (PRC1 and PRC2) have been shown to inhibit reprogramming. Repression of histone methyl transferases SUV39H1[61] and Dot1L[62] as well as YY1[63] enhanced reprogramming. Specifically, repression of the catalytic activity Dot1L reduced H3K79 methylation and increased reprogramming efficiency by 3-4 fold[49]. DNA methylation and demethylation are crucial bottlenecks during reprogramming, but because of the complexities of the signals and their importance to appropriate gene expression, in looking for effective enhancers of reprograming, targeted inhibition of specific methylation inhibitors may give a better results.

KINASES

The focus of understanding the mechanisms underlying reprogramming have typically concentrated on transcription factors and chromatin modifying factors. However, inhibition of select kinases has also been shown to increase reprogramming efficiency[13,64]. Examples include the TGF-βr1 and SRC kinases, although only inhibition of TGF-βr1 was able to enhance reprogramming in the absence of VPA[13]. Inhibition of TGF-βr1 allowed reprogramming to occur in the absence of Sox2 by inducing Nanog expression in intermediately reprogrammed cells that had been exposed to Oct4, Klf4 and c-Myc[13]. A small chemical screen identified additional kinase barriers to reprogramming that include the MAP p38 kinase[65], IP3K, which catalyzes synthesis of a crucial secondary lipid messenger required for Ca+ signaling[66], and Aurora A kinase, which is essential for cell division[67]. Inhibition of each of these enhanced iPSC cell formation, indicating that factors that may have more selected specificities beyond the transcription factors that activate them may be worthy factors for inhibition.

CONCLUSION

The potential of human embryonic stem (hESC) cells in the clinic has been understood long before science was able to deliver on that potential. The issues that may limit using hESC as a therapeutic, such as potential immune rejection and ethical concerns, has made iPSC cells a very attractive alternative approach. iPSC appear to have the same ability as hESC cells to differentiate in vitro and so it is hoped that they will be able to replace hESC cells therapeutically. The underlying problem is efficient creation of reliable, stable iPSC without gene integration[68-71]. While there have been massive strides made in the field of reprogramming, there is still a lack of clinically useful methods for producing iPSC that have been shown to be safe in the long term[16,72-74]. Future work must expand on creating more robust reprogramming protocols, rather than staying limited to established reprogramming methods. We must look at all the proteins and microRNAs in the cell, not just known transcription factors. We must look at both enhancers of pluripotency and repressors of pluripotency as possible targets. The field has advanced enormously in only a few short years; however in order to fully manipulate cell fate, we must be more inclusive when searching for cellular reprogramming factors and broaden our understanding of how cell fate is determined.

Footnotes

P- Reviewer: Islam MQ, Pantakani DVK S- Editor: Gong XM L- Editor: A E- Editor: Lu YJ

References
1.  Gurdon JB. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J Embryol Exp Morphol. 1962;10:622-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
2.  Gurdon JB. The developmental capacity of nuclei taken from differentiating endoderm cells of Xenopus laevis. J Embryol Exp Morphol. 1960;8:505-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
3.  Campbell KH, McWhir J, Ritchie WA, Wilmut I. Sheep cloned by nuclear transfer from a cultured cell line. Nature. 1996;380:64-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1280]  [Cited by in F6Publishing: 1034]  [Article Influence: 36.9]  [Reference Citation Analysis (0)]
4.  Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mammalian cells. Cloning Stem Cells. 2007;9:3-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3629]  [Cited by in F6Publishing: 2931]  [Article Influence: 108.6]  [Reference Citation Analysis (0)]
5.  Tachibana M, Amato P, Sparman M, Gutierrez NM, Tippner-Hedges R, Ma H, Kang E, Fulati A, Lee HS, Sritanaudomchai H. Human embryonic stem cells derived by somatic cell nuclear transfer. Cell. 2013;153:1228-1238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 552]  [Cited by in F6Publishing: 471]  [Article Influence: 42.8]  [Reference Citation Analysis (0)]
6.  Chung YG, Eum JH, Lee JE, Shim SH, Sepilian V, Hong SW, Lee Y, Treff NR, Choi YH, Kimbrel EA. Human somatic cell nuclear transfer using adult cells. Cell Stem Cell. 2014;14:777-780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 142]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
7.  Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17989]  [Cited by in F6Publishing: 17070]  [Article Influence: 948.3]  [Reference Citation Analysis (0)]
8.  Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14327]  [Cited by in F6Publishing: 13579]  [Article Influence: 848.7]  [Reference Citation Analysis (0)]
9.  Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, Thomson JA. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324:797-801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1735]  [Cited by in F6Publishing: 1628]  [Article Influence: 108.5]  [Reference Citation Analysis (0)]
10.  Lin T, Ambasudhan R, Yuan X, Li W, Hilcove S, Abujarour R, Lin X, Hahm HS, Hao E, Hayek A. A chemical platform for improved induction of human iPSCs. Nat Methods. 2009;6:805-808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 460]  [Cited by in F6Publishing: 418]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
11.  Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE, Melton DA. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol. 2008;26:795-797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1249]  [Cited by in F6Publishing: 1170]  [Article Influence: 73.1]  [Reference Citation Analysis (0)]
12.  Feng B, Ng JH, Heng JC, Ng HH. Molecules that promote or enhance reprogramming of somatic cells to induced pluripotent stem cells. Cell Stem Cell. 2009;4:301-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 265]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
13.  Ichida JK, Blanchard J, Lam K, Son EY, Chung JE, Egli D, Loh KM, Carter AC, Di Giorgio FP, Koszka K. A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell. 2009;5:491-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 632]  [Cited by in F6Publishing: 594]  [Article Influence: 39.6]  [Reference Citation Analysis (0)]
14.  Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, Zhang Y, Yang W, Gruber PJ, Epstein JA. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 2011;8:376-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 900]  [Cited by in F6Publishing: 869]  [Article Influence: 66.8]  [Reference Citation Analysis (0)]
15.  Hussein SM, Nagy AA. Progress made in the reprogramming field: new factors, new strategies and a new outlook. Curr Opin Genet Dev. 2012;22:435-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
16.  Ma H, Morey R, O’Neil RC, He Y, Daughtry B, Schultz MD, Hariharan M, Nery JR, Castanon R, Sabatini K. Abnormalities in human pluripotent cells due to reprogramming mechanisms. Nature. 2014;511:177-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 251]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
17.  Zhao Y, Yin X, Qin H, Zhu F, Liu H, Yang W, Zhang Q, Xiang C, Hou P, Song Z. Two supporting factors greatly improve the efficiency of human iPSC generation. Cell Stem Cell. 2008;3:475-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 343]  [Article Influence: 22.9]  [Reference Citation Analysis (0)]
18.  Sarig R, Rivlin N, Brosh R, Bornstein C, Kamer I, Ezra O, Molchadsky A, Goldfinger N, Brenner O, Rotter V. Mutant p53 facilitates somatic cell reprogramming and augments the malignant potential of reprogrammed cells. J Exp Med. 2010;207:2127-2140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 123]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
19.  Utikal J, Polo JM, Stadtfeld M, Maherali N, Kulalert W, Walsh RM, Khalil A, Rheinwald JG, Hochedlinger K. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature. 2009;460:1145-1148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 654]  [Cited by in F6Publishing: 648]  [Article Influence: 43.2]  [Reference Citation Analysis (0)]
20.  Marión RM, Strati K, Li H, Murga M, Blanco R, Ortega S, Fernandez-Capetillo O, Serrano M, Blasco MA. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature. 2009;460:1149-1153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 785]  [Cited by in F6Publishing: 789]  [Article Influence: 52.6]  [Reference Citation Analysis (0)]
21.  Li H, Collado M, Villasante A, Strati K, Ortega S, Cañamero M, Blasco MA, Serrano M. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature. 2009;460:1136-1139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 739]  [Cited by in F6Publishing: 744]  [Article Influence: 49.6]  [Reference Citation Analysis (0)]
22.  Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, Raya A, Wahl GM, Izpisúa Belmonte JC. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature. 2009;460:1140-1144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 841]  [Cited by in F6Publishing: 854]  [Article Influence: 56.9]  [Reference Citation Analysis (0)]
23.  Hanna J, Saha K, Pando B, van Zon J, Lengner CJ, Creyghton MP, van Oudenaarden A, Jaenisch R. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature. 2009;462:595-601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 797]  [Cited by in F6Publishing: 735]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
24.  Meek DW. Tumour suppression by p53: a role for the DNA damage response? Nat Rev Cancer. 2009;9:714-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 435]  [Cited by in F6Publishing: 457]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
25.  Brosh R, Rotter V. When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer. 2009;9:701-713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 811]  [Cited by in F6Publishing: 831]  [Article Influence: 55.4]  [Reference Citation Analysis (0)]
26.  Vousden KH, Ryan KM. p53 and metabolism. Nat Rev Cancer. 2009;9:691-700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 708]  [Cited by in F6Publishing: 717]  [Article Influence: 47.8]  [Reference Citation Analysis (0)]
27.  Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6844]  [Cited by in F6Publishing: 6706]  [Article Influence: 291.6]  [Reference Citation Analysis (0)]
28.  Tung PY, Knoepfler PS. Epigenetic mechanisms of tumorigenicity manifesting in stem cells. Oncogene. 2014;Jun 16; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
29.  Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa M, Okita K, Yamanaka S. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature. 2009;460:1132-1135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1004]  [Cited by in F6Publishing: 996]  [Article Influence: 66.4]  [Reference Citation Analysis (0)]
30.  Popowski M, Templeton TD, Lee BK, Rhee C, Li H, Miner C, Dekker JD, Orlanski S, Bergman Y, Iyer VR. Bright/Arid3A acts as a barrier to somatic cell reprogramming through direct regulation of Oct4, Sox2, and Nanog. Stem Cell Reports. 2014;2:26-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
31.  Wang J, Rao S, Chu J, Shen X, Levasseur DN, Theunissen TW, Orkin SH. A protein interaction network for pluripotency of embryonic stem cells. Nature. 2006;444:364-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 849]  [Cited by in F6Publishing: 840]  [Article Influence: 46.7]  [Reference Citation Analysis (0)]
32.  Herrscher RF, Kaplan MH, Lelsz DL, Das C, Scheuermann R, Tucker PW. The immunoglobulin heavy-chain matrix-associating regions are bound by Bright: a B cell-specific trans-activator that describes a new DNA-binding protein family. Genes Dev. 1995;9:3067-3082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 211]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
33.  Lin D, Ippolito GC, Zong RT, Bryant J, Koslovsky J, Tucker P. Bright/ARID3A contributes to chromatin accessibility of the immunoglobulin heavy chain enhancer. Mol Cancer. 2007;6:23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
34.  Kortschak RD, Tucker PW, Saint R. ARID proteins come in from the desert. Trends Biochem Sci. 2000;25:294-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 136]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
35.  Webb CF, Bryant J, Popowski M, Allred L, Kim D, Harriss J, Schmidt C, Miner CA, Rose K, Cheng HL. The ARID family transcription factor bright is required for both hematopoietic stem cell and B lineage development. Mol Cell Biol. 2011;31:1041-1053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
36.  Kaiwen M. [Involvement of E2FBP1, an ARID family member protein, in the p53 regulatory pathway]. Kokubyo Gakkai Zasshi. 2002;69:152-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
37.  Ma K, Araki K, Ichwan SJ, Suganuma T, Tamamori-Adachi M, Ikeda MA. E2FBP1/DRIL1, an AT-rich interaction domain-family transcription factor, is regulated by p53. Mol Cancer Res. 2003;1:438-444.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  An G, Miner CA, Nixon JC, Kincade PW, Bryant J, Tucker PW, Webb CF. Loss of Bright/ARID3a function promotes developmental plasticity. Stem Cells. 2010;28:1560-1567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 36]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
39.  Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science. 2001;293:1089-1093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2232]  [Cited by in F6Publishing: 2038]  [Article Influence: 88.6]  [Reference Citation Analysis (0)]
40.  Lee JH, Hart SR, Skalnik DG. Histone deacetylase activity is required for embryonic stem cell differentiation. Genesis. 2004;38:32-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 203]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
41.  Pasini D, Bracken AP, Hansen JB, Capillo M, Helin K. The polycomb group protein Suz12 is required for embryonic stem cell differentiation. Mol Cell Biol. 2007;27:3769-3779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 535]  [Cited by in F6Publishing: 532]  [Article Influence: 31.3]  [Reference Citation Analysis (0)]
42.  Peters AH, Kubicek S, Mechtler K, O’Sullivan RJ, Derijck AA, Perez-Burgos L, Kohlmaier A, Opravil S, Tachibana M, Shinkai Y. Partitioning and plasticity of repressive histone methylation states in mammalian chromatin. Mol Cell. 2003;12:1577-1589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 843]  [Cited by in F6Publishing: 847]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
43.  Rice JC, Briggs SD, Ueberheide B, Barber CM, Shabanowitz J, Hunt DF, Shinkai Y, Allis CD. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol Cell. 2003;12:1591-1598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 591]  [Cited by in F6Publishing: 590]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
44.  Tachibana M, Ueda J, Fukuda M, Takeda N, Ohta T, Iwanari H, Sakihama T, Kodama T, Hamakubo T, Shinkai Y. Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3-K9. Genes Dev. 2005;19:815-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 600]  [Cited by in F6Publishing: 593]  [Article Influence: 31.2]  [Reference Citation Analysis (0)]
45.  Eissenberg JC, Shilatifard A. Histone H3 lysine 4 (H3K4) methylation in development and differentiation. Dev Biol. 2010;339:240-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 228]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
46.  South PF, Harmeyer KM, Serratore ND, Briggs SD. H3K4 methyltransferase Set1 is involved in maintenance of ergosterol homeostasis and resistance to Brefeldin A. Proc Natl Acad Sci USA. 2013;110:E1016-E1025.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
47.  Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39812]  [Cited by in F6Publishing: 43087]  [Article Influence: 3314.4]  [Reference Citation Analysis (4)]
48.  Buganim Y, Faddah DA, Jaenisch R. Mechanisms and models of somatic cell reprogramming. Nat Rev Genet. 2013;14:427-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 333]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
49.  Onder TT, Kara N, Cherry A, Sinha AU, Zhu N, Bernt KM, Cahan P, Marcarci BO, Unternaehrer J, Gupta PB. Chromatin-modifying enzymes as modulators of reprogramming. Nature. 2012;483:598-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 505]  [Cited by in F6Publishing: 488]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
50.  Soufi A, Donahue G, Zaret KS. Facilitators and impediments of the pluripotency reprogramming factors’ initial engagement with the genome. Cell. 2012;151:994-1004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 717]  [Cited by in F6Publishing: 643]  [Article Influence: 53.6]  [Reference Citation Analysis (0)]
51.  Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 2006;125:315-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3971]  [Cited by in F6Publishing: 3891]  [Article Influence: 216.2]  [Reference Citation Analysis (0)]
52.  Marks H, Kalkan T, Menafra R, Denissov S, Jones K, Hofemeister H, Nichols J, Kranz A, Stewart AF, Smith A. The transcriptional and epigenomic foundations of ground state pluripotency. Cell. 2012;149:590-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 681]  [Cited by in F6Publishing: 628]  [Article Influence: 52.3]  [Reference Citation Analysis (0)]
53.  Kretsovali A, Hadjimichael C, Charmpilas N. Histone deacetylase inhibitors in cell pluripotency, differentiation, and reprogramming. Stem Cells Int. 2012;2012:184154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 88]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
54.  Göttlicher M, Minucci S, Zhu P, Krämer OH, Schimpf A, Giavara S, Sleeman JP, Lo Coco F, Nervi C, Pelicci PG. Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J. 2001;20:6969-6978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1375]  [Cited by in F6Publishing: 1404]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
55.  Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, Chen S, Muhlestein W, Melton DA. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat Biotechnol. 2008;26:1269-1275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 994]  [Cited by in F6Publishing: 922]  [Article Influence: 57.6]  [Reference Citation Analysis (0)]
56.  Zhou H, Wu S, Joo JY, Zhu S, Han DW, Lin T, Trauger S, Bien G, Yao S, Zhu Y. Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell. 2009;4:381-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1309]  [Cited by in F6Publishing: 1208]  [Article Influence: 80.5]  [Reference Citation Analysis (0)]
57.  Planello AC, Ji J, Sharma V, Singhania R, Mbabaali F, Müller F, Alfaro JA, Bock C, De Carvalho DD, Batada NN. Aberrant DNA methylation reprogramming during induced pluripotent stem cell generation is dependent on the choice of reprogramming factors. Cell Regen (Lond). 2014;3:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
58.  Chiu CP, Blau HM. 5-Azacytidine permits gene activation in a previously noninducible cell type. Cell. 1985;40:417-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 84]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
59.  Bhutani N, Brady JJ, Damian M, Sacco A, Corbel SY, Blau HM. Reprogramming towards pluripotency requires AID-dependent DNA demethylation. Nature. 2010;463:1042-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 526]  [Cited by in F6Publishing: 512]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
60.  Deng W. AID in reprogramming: quick and efficient: identification of a key enzyme called AID, and its activity in DNA demethylation, may help to overcome a pivotal epigenetic barrier in reprogramming somatic cells toward pluripotency. Bioessays. 2010;32:385-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
61.  Schotta G, Ebert A, Reuter G. SU(VAR)3-9 is a conserved key function in heterochromatic gene silencing. Genetica. 2003;117:149-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 496]  [Cited by in F6Publishing: 484]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
62.  Jones B, Su H, Bhat A, Lei H, Bajko J, Hevi S, Baltus GA, Kadam S, Zhai H, Valdez R. The histone H3K79 methyltransferase Dot1L is essential for mammalian development and heterochromatin structure. PLoS Genet. 2008;4:e1000190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 270]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
63.  Shi Y, Seto E, Chang LS, Shenk T. Transcriptional repression by YY1, a human GLI-Krüppel-related protein, and relief of repression by adenovirus E1A protein. Cell. 1991;67:377-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 740]  [Cited by in F6Publishing: 830]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
64.  Li Z, Rana TM. A kinase inhibitor screen identifies small-molecule enhancers of reprogramming and iPS cell generation. Nat Commun. 2012;3:1085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 79]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
65.  Zarubin T, Han J. Activation and signaling of the p38 MAP kinase pathway. Cell Res. 2005;15:11-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1091]  [Cited by in F6Publishing: 1149]  [Article Influence: 60.5]  [Reference Citation Analysis (0)]
66.  Xia HJ, Yang G. Inositol 1,4,5-trisphosphate 3-kinases: functions and regulations. Cell Res. 2005;15:83-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 50]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
67.  Kollareddy M, Zheleva D, Dzubak P, Brahmkshatriya PS, Lepsik M, Hajduch M. Aurora kinase inhibitors: progress towards the clinic. Invest New Drugs. 2012;30:2411-2432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 179]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
68.  Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008;322:949-953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1459]  [Cited by in F6Publishing: 1301]  [Article Influence: 81.3]  [Reference Citation Analysis (0)]
69.  Carey BW, Markoulaki S, Hanna J, Saha K, Gao Q, Mitalipova M, Jaenisch R. Reprogramming of murine and human somatic cells using a single polycistronic vector. Proc Natl Acad Sci USA. 2009;106:157-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 387]  [Cited by in F6Publishing: 397]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
70.  Gonzalez F, Barragan Monasterio M, Tiscornia G, Montserrat Pulido N, Vassena R, Batlle Morera L, Rodriguez Piza I, Izpisua Belmonte JC. Generation of mouse-induced pluripotent stem cells by transient expression of a single nonviral polycistronic vector. Proc Natl Acad Sci USA. 2009;106:8918-8922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 205]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
71.  Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P, Woltjen K. Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature. 2009;458:771-775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 942]  [Cited by in F6Publishing: 843]  [Article Influence: 56.2]  [Reference Citation Analysis (0)]
72.  Hussein SM, Batada NN, Vuoristo S, Ching RW, Autio R, Närvä E, Ng S, Sourour M, Hämäläinen R, Olsson C. Copy number variation and selection during reprogramming to pluripotency. Nature. 2011;471:58-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 722]  [Cited by in F6Publishing: 681]  [Article Influence: 52.4]  [Reference Citation Analysis (0)]
73.  Gore A, Li Z, Fung HL, Young JE, Agarwal S, Antosiewicz-Bourget J, Canto I, Giorgetti A, Israel MA, Kiskinis E. Somatic coding mutations in human induced pluripotent stem cells. Nature. 2011;471:63-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 956]  [Cited by in F6Publishing: 922]  [Article Influence: 70.9]  [Reference Citation Analysis (0)]
74.  Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR, Hon G, Antosiewicz-Bourget J, O’Malley R, Castanon R, Klugman S. Hotspots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature. 2011;471:68-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1202]  [Cited by in F6Publishing: 1114]  [Article Influence: 85.7]  [Reference Citation Analysis (0)]