Available online at www.sciencedirect.com

ScienceDirect Cellular reprogramming by transcription factor engineering Jason CH Tsang1, Xuefei Gao1, Liming Lu1,3 and Pentao Liu1,2 Recent researches have identified multiple transcription factors as permissible reprogramming factors to pluripotency and lineage switching. The current standard strategy by ectopic factor overexpression however has intrinsic limitations in studying the reprogramming mechanism. There is a growing interest in engineering novel chimeric reprogramming factors and applying designer transcription factors technology to improve reprogramming efficiency and dissect the process of endogenous pluripotency network reactivation. Here, we provide a concise review on the latest progress in studying cellular reprogramming by transcription factor engineering. Addresses 1 Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK 2 Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Rd, Cambridge CB2 1QR, UK 3 Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, 200025, China Corresponding authors: Lu, Liming ([email protected]) and Liu, Pentao ([email protected])

somatic lineage switching by ectopic expression of transcription factor CEBPA and MYOD [3,4]. These works highlighted the fact that reprogramming factors are often the master transcription factors that govern the epigenetic state and the cell identity. Such realization laid the ground for Takahashi and Yamanaka’s landmark discovery of pluripotency transcription factors, Oct4, Sox2, Klf4 and c-Myc (OSKM), as defined reprogramming factors to pluripotency [5]. This set of factor is conserved and applicable to virtually all somatic cell types [6–12]. The simplicity and clinical potential of reprogramming to pluripotency fuelled a flurry of search for new reprogramming factors. Genome-wide genetic screening later uncovered surprising reprogramming capabilities of many candidates [13]. Currently, dozens of genetic factors have been reported to enhance or substitute OSKM in the process of reprogramming to pluripotency upon genetic perturbation. Although chemical-mediated reprogramming showed significant promise [14], the most common approach remains ectopic genetic factor expression.

Current Opinion in Genetics & Development 2014, 28:1–9 This review comes from a themed issue on Cell reprogramming, regeneration and repair Edited by Jose´ CR Silva and Renee A Reijo Pera

http://dx.doi.org/10.1016/j.gde.2014.07.001 0959-437X/# 2014 Elsevier Ltd. All right reserved.

Transcription factor emerges as the key to reprogramming The current paradigm in developmental biology supports a model of epigenesis, where development follows a hierarchical cell fate decision from the zygote. It has been long held that the loss of developmental potency is only reversible in germ cells after fertilization as a natural form of cellular reprogramming. It is later demonstrated that somatic nucleus is also plastic to reprogramming by artificial somatic nuclear transfer (SCNT) and it provided the basis of the existence of reprogramming factors in the oocyte cytoplasm [1]. Embryonic stem cells were also shown to contain reprogramming factors that can induce pluripotency from somatic cells [2]. It is corroborated by early reports of www.sciencedirect.com

Limitations of ectopic genetic factor overexpression in reprogramming In spite of the success, expressing exogenous genetic factors has intrinsic limitations. Epigenetic reprogramming is an artificial in vitro process and native genetic factors are not originally evolved for such purpose. Their operational concentrations and partners are normally highly regulated under physiological conditions [15,16]. Caution is needed to interpret their molecular behaviour during reprogramming as an extension of their native function. Multiple lines of evidence suggested that somatic reprogramming by ectopic factor overexpression follows a defined molecular roadmap and the reprogramming factors coordinate to reactivate the endogenous pluripotency network [17–19]. Genomewide profiling of reprogramming factor DNA binding at early stage of reprogramming reveals significant nonspecific interaction and aberrant activation of somatic and apoptotic program [17,20]. Importantly, large pluripotency-associated genomic regions, for example, the Sox2 and Nanog loci, are often refractory to reactivation, resulting in stalling and the stochastic nature of reprogramming. Somatic reprogramming by ectopic factor overexpression is also sensitive to vector design and expression stoichiometry. High Sox2 expression is detrimental and low Oct4 and Klf4 have been associated with abnormal imprinting in iPS cells [21–24]. Current Opinion in Genetics & Development 2014, 28:1–9

2 Cell reprogramming, regeneration and repair

Improving reprogramming by native transcription factor engineering In view of these shortcomings, there is growing interest to explore alternative reprogramming approaches such as engineering transcription factors to augment reprogramming potency and reactivation specificity. The capability of somatic reprogramming to pluripotency by OSKM is conserved in vertebrates. It is therefore intuitive to seek inspiration from the natural variation of OSKM from the phylogenetic tree. One study specifically looked at the conservation of Nanog in vertebrates and surprisingly they reported marked efficiency improvement in mouse with chick and zebrafish Nanog [25]. Systematic evaluation of inter-species variation and reprogramming potency of OSKM is however scarce. Although paralogues of individual gene family of OSKM showed high sequence homology, the capability of mediating efficient somatic reprogramming appears to be unique to OSKM with an exception of Klf2, which marginally outperformed Klf4 [26]. It is suggested that such specificity is exerted through selective interaction between reprogramming factors, which in turn is determined by the unique interaction interface of each member [27,28]. It is supported by studies of native transcription factor engineering. Subtle changes in the coding sequence at specific domain can confer new reprogramming ability to previously impermissible factors or abolish the ability of existing one. SOX17 can be converted to a reprogramming factor by exchanging a single amino acid at the OCT4 interaction domain with SOX2 [28]. This re-engineered SOX17 is three times more efficient than native SOX2 in reprogramming. Moreover, this study also established the essentiality of cooperative action of OSKM in reprogramming. Rational engineering or random mutagenesis study of the coding sequence of OSKM may further discover new variants and yield better understanding on reprogramming factor biochemistry. As OCT4, SOX2 and KLF4 all possess a modular protein structure with defined DNA binding and effector protein domains, larger scale of modular engineering is feasible. Several groups have generated chimeric reprogramming factors by replacing the effector domain of OCT4 with alternative effector domain such as the transactivation domain of VP16, MYOD and YAP [29,30,31]. These chimeric factors enhanced recruitment of histone modifiers p300 and their interaction with demethylation enzyme TET1/2, thus drastically accelerate epigenetic reprogramming.

Dissecting reprogramming by designer transcription factor engineering The zinc finger and TAL effector technology

Another approach that gained significant momentum recently is designer transcription factor engineering. It is an expansion of the concept of modular engineering to generate site-specific transcription factors to control transcription of selected genomic region. It has the potential to target specific roadblocks in reprogramming and reactivation of pluripotency-associated loci. Various chemical Current Opinion in Genetics & Development 2014, 28:1–9

approaches have been successfully applied to create artificial transcription factors to modulate gene expression [32]. In the following part of the review, we focused the discussion on the latest development of protein-based approach and their relevance to cellular reprogramming. The ‘Zinc Finger Protein’ (ZFP) technology is the prototype platform of this kind and is pioneered in the 1990s after the structure of the zinc finger motif was resolved [33]. The DNA binding region of zinc finger protein consists of an array of Cys2-His2 zinc fingers. Each zinc finger recognizes 3 bp of DNA. A site-specific designer transcription factor can be generated by fusing effector domains to a custom-made zinc fingers array [34]. Effector-domain-free ZFP can also modulate gene expression by blocking native transcription factor binding [35]. This approach has achieved successes in modulating OSKM expression and inducing ES cell differentiation [36,37]. Nonetheless, popularization of the platform remained limited. A number of resources have been built to simplify the screening and assembly workflow [38,39,40], but adoption is still slow. This barrier has been recently overcome by the introduction of the ‘Transcription Activator-like Effector’ (TALE) technology. In principle, it is a succession of the ZFP platform. TALEs are natural transcription factor produced by pathogen Xanthomonas to modulate gene expression of their plant host. The DNA binding domain of TALE has a repeat structure and each repeat differs at the 12th and 13th amino acid position (the Repeat Variable Diresidue, RVD). It is later found that this peculiar configuration encodes the DNA binding specificity with a simple 1:1 repeat-to-nucleotide recognition logic [41,42]. Therefore, a TALE DNA binding domain with good specificity can be easily synthesized by assembling a TALE repeat array with corresponding RVDs. Different high-throughput TALE assembly platforms have been developed to streamline the cloning process [43–47]. With the improved scalability, synthetic TALE can now be used to systematically interrogate different genomic regions and study their effect on transcription modulation and biological consequence in detail. Similar to ZF effectors, TALEs are able to modulate endogenous Oct4 expression in ES cells and reactivate Oct4 expression in somatic cells [48,49,50]. Our group exploited the strength of the TALE technology and went further from gene activation to study the process of somatic reprogramming and revealed three interesting observations [51]. Firstly, we demonstrated the feasibility of replacing ectopic Oct4 and Nanog overexpression and instructing reprogramming by designer TALE transcription factor targeting the Oct4 and Nanog enhancers. Secondly, we observed that TALE mediated reprogramming displays a different reprogramming kinetics. In MEF reprogramming, the endogenous Oct4 is readily reactivated by TALE at day 5 with a linear kinetics, whereas reactivation by ectopic expression is initially absent until day 9 of induction with an exponential burst. Unexpectedly www.sciencedirect.com

Creating the ‘‘Swiss Knives’’ for reprogramming Tsang et al.

though, the time point of endogenous Rex1 reactivation remains similar and the overall efficiency is actually lower than ectopic expression, indicating that early reactivation of Oct4 alone is not sufficient. It is worthy noting that the combination of the two approaches has synergistic effect in reprogramming (Figure 1). Thirdly, the genetic context of the targeting site also influences reprogramming. Gene promoters interact with transcription initiation complex to trigger mRNA transcription, therefore most studies target TALEs to the promoter for transcription modulation. Meanwhile, the importance of other genetic elements in gene transcription such as enhancers is increasingly recognized [52,53]. It is illustrated by our finding that enhancer TALEs is more efficient than promoter TALEs in MEF reprogramming [51]. This is actually not unexpected, as previous work studying the mechanism of reprogramming initiation at the Myod1 locus indicates that OCT4 and MYOD regulate Myod1

3

expression through interaction with permissible enhancers first before inducing epigenetic changes at the promoter [54]. Moreover, the Oct4 distal enhancer has been shown to be the most densely bound genomic region by multiple pluripotency transcription factors, like SOX2, MYC and KLF4, and signalling transducer STAT3 and SMAD1 in ES cells [55], targeted activation by TALEs may have broad facilitating effect on reactivating the pluripotency network. Given the high tissue-specific activity and the latest discovery of large tissue-specific enhancer clusters [56,57], enhancer manipulation by designer transcription factor may hold the key to dissect the mechanism of tissue-specific gene network reactivation in reprogramming and development. Attempt to manipulate developmental specific enhancers in vivo has been successful in Drosophila [58]. It awaits to be shown that enhancer TALEs can completely replace exogenous factors in mediating reprogramming.

Figure 1

The Silenced Oct4 (Pou5f1) locus in MEF

K

Exogenous factor

K

Endogenous factor H3K27me3

Pou5f1

Distal Enhancer (DE)

H3K27Ac H3K4me3

Proximal Promoter (PP)

1. Standard OSKM induction

M

2. TALE activator-assisted OSKM induction p300

p300

K

Omitted segment

Epigenetic modifiers recruitment

+

PolII

O S

p300

p300

K

Pou5f1

DE

M

PolII

O S

Pou5f1

DE

O S

O S

PP

PP Enhancer initiated promoter reactivation S K M O

S K M O

p300

OS

p300 p300V

Pou5f1 PolII

PolII

O S

N K

PolII

Pou5f1 PolII

T S M O

L p300 Mediator L

PolII

PolII

Pou5f1

Successful reprogramming Current Opinion in Genetics & Development

A schematic diagram showing the reactivation of the Oct4 locus by standard exogenous transcription factor and designer transcription factor in reprogramming. Designer transcription factors improve reprogramming efficiency by enhancing recruitment of epigenetic modifiers to the Oct4 enhancers and thus assisting reactivation of the Oct4 promoter. The filled circles depict the epigenetic histone modifications. O: OCT4; K: KLF4; S: SOX2; M: MYC; L: LRH1; T: TCFCP2L1. www.sciencedirect.com

Current Opinion in Genetics & Development 2014, 28:1–9

4 Cell reprogramming, regeneration and repair

In additions to gene reactivation, another study reported the use of TALEs to repress the pluripotency-associated microRNA-302/367 clusters in fibroblast and successfully impede somatic reprogramming [59]. This study highlighted several advantages of designer transcription factor in gene repression. Compared to standard RNA interference, designer transcription factors repress gene expression at the transcription level. It is particularly useful for small RNA repression, as they are often transcribed in clusters and prone to interference saturation. This tool will help uncovering the role of microRNAs in reprogramming [60,61]. Depending on the molecular characteristics of the repressor domain, the reversibility of the repression can vary. The initial repression can be reversible due to direct interference with the transcription machinery, but permanent silencing can be induced by recruitment of repressive complex and deposition of stable epigenetic changes such as promoter methylation. It will be interesting to compare the phenotypes of lossof-function experiments from post-transcriptional depletion by RNAi, transcriptional repression by designer TALE repressor and genetic deletion by mutagenic knockout. Furthermore, an optogenetic TALE system has been developed to permit precise temporal and spatial rewriting of the regional histone code by fusing TALE to methyltransferase and deacetylase domains [62]. Specific technical optimizations such as the use of valporic acid to assist gene reactivation and characterization of new RVDs with higher affinity to methylated cytosine have also been explored [49,63,64]. The CRISPR technology

The versatility of the TALE system has tremendously enriched the repertoire of biologist to investigate complex biological problems, but the even more exciting progress in designer transcription factor engineering is indeed the latest innovation of the RNA-guided Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) system. CRISPR is originally a natural adaptive immune system in bacteria against virus infection [65]. The viral DNA sequence is integrated into the cas operon to generate an array of proto-spacers. Upon reinfection, the cas operon produces two non-coding RNAs: the trans-activating CRISPR RNA (tracrRNA) and the precursor CRISPR RNA (pre-crRNA). The later contains the guide spacer sequence (20 bp). The two RNAs can direct the nuclease Cas9 to cleave the foreign DNA by complementary RNA–DNA pairing of the target phage DNA and the spacer RNA sequence. The system is later simplified and adapted for genome editing in mammalian cells [66–69], it has also been repurposed to create a RNA-guided designer transcription factor system by inactivating the Cas9 nuclease function (dCas9) [70]. Similar to the TALE technology, various effector domains can be fused to the deactivated Cas9 protein [71,72,73]. The CRISPR system provides a further growth in throughput compared to the TALE system due to the superior Current Opinion in Genetics & Development 2014, 28:1–9

scalability and economy of oligonucleotide synthesis to modular TALE protein assembly. The CRISPR system is also highly multiplexable, multiple guide RNAs can be delivered and can modulate multiple loci simultaneously [74]. Nonetheless, the TALE system has the advantage of customizable TALE domain length and less restriction in target sequence selection. Paralleled activation and repression is also practical by TALE. The modulating efficiency of the two systems however has not been formally compared. Success of guiding cellular differentiation in human ES cells by the CRISPR system has been recently reported [75]. Yet, the suitability of the CRISPR system in reprogramming remains to be investigated. One distinguishing feature of these two systems is the molecular mechanism in target DNA sequence recognition. The TALE domain wraps around the major groove of the target DNA to form a superhelix, while the single guide RNA (sgRNA) forms a heteroduplex with the target DNA strand in the CRISPR system [76–78]. This difference may have consequences in transcription modulation as RNA/DNA heteroduplex may interfere with native transcription factor and RNA polymerase binding at neighbouring regions. This phenomenon, known as CRISPR interference, was first reported at gene promoter and later adapted as a means to repress gene expression [70]. However, the effect of interference may be context dependent [75]. Another aspect demanding caution is the possible inadvertent mutagenesis due to the residual nuclease activity and non-specific targeting of the RNA/DNA pairing [79–81]. A recent study investigating the genome-wide binding of the dCas9/sgRNA complex revealed that DNA binding is only dependent on a short pentameric seed region and the NGG protospacer adjacent motif (PAM) of the sgRNA, however, successful DNA cleavage by Cas9 requires more extensive complementary sgRNA/DNA pairing [82]. Therefore, it can be over-optimistic to directly extrapolate the high mutagenic specificity of the CRISPR system to gene modulating applications. These issues are of special significance in long-term experiment such as reprogramming and development, which require prolonged expression of CRISPR effectors. In spite of these, the flexibility of design has made these two systems the ‘Swiss knives’ in gene regulation.

The future and beyond In summary, transcription factor engineering has found promising applications in dissecting the mechanism and improving the efficiency of reprogramming. It is a unique strategy that is at the intersection of our knowledge on protein biochemistry, genome biology and the latest innovation in biotechnology. The development of designer transcription factor engineering in the last five years has been breath taking. A major technical hurdle of synthesis throughput has been overcome by the introduction of the next-generation TALE and CRISPR platforms. With these valuable tools in hands, biologists are www.sciencedirect.com

Creating the ‘‘Swiss Knives’’ for reprogramming Tsang et al.

now able to examine important issues in somatic reprogramming and lineage specification in a fresh perspective such as identification of the key factors in endogenous pluripotency network reactivation and the role of different genetic regulatory elements in reprogramming and differentiation (Figure 2). Large-scale application such as regulatory element and gene perturbation screening by CRISPR effectors is now realistic with genome-wide sgRNA library and TALE library screening has also been reported [83]. This offers a complementary strategy for novel reprogramming factor or roadblock discovery and tissue-specific enhancer validation to the standard RNAi knockdown and mutagenesis approach [84–86]. The chromatin conformation of the Nanog locus has recently been elucidated and the change of organization has been shown to

5

be critical in reprogramming [87,88]. Meanwhile, long-range interaction manipulation of the locus-controlling region and b-globin gene has been demonstrated by fusing respective ZFP and looping factor LDB1 [89]. It is therefore foreseeable that higher order manipulation at multiple loci by TALE and CRISPR effectors may be used to study reprogramming in the future. Other creative applications of the TALE and CRISPR platforms involve locus tracking by tagging locus-specific effectors with fluorescent proteins and site-specific cassette insertion with transposase fusion [90,91] (Figure 3). Additional potential applications of designer transcription factors including functional validation of non-coding variants identified by population genetic studies and controlling expression of genetic isoforms by alternative promoter modulation remains to be explored.

Figure 2

(a) Direct Activation of Lineage Specifier

M

Target 1

M

Target 2

(b) Indirect Activation of Lineage Specifier through Network Enhancer Lineage A Program PolII

M

+

PolII

Target 3

Progenitors Stem Cells

PolII Master Regulator

Gene specific promoter/ enhancer

Target A1 P1

M M2 M1 + p300 M3

Target A2 P2

PolII

Locus controlling region “Enhanceosome”

Target A3 P3

PolII

Reprogramming Differentiation

Target A4 P4

PolII

Lineage A Program +

Lineage A

PolII

Target A5 P5

Lineage B

Target A1 P1

+

Lineage A Program

PolII Target A2 P2

+

+

PolII Target A3 P3

+

PolII Target A4 PolII

Gene specific promoter



MA

Master Regulator A

P2

MA MB

PolII

P1

Target 1 Target 2

Lineage B Program

Master Regulator B

P4

+

PolII

Target A5 P5

(c) Direct Activation of Lineage Program through Multiplexed Activators

Gene specific promoter

P3 P4

Target 3 Target 4

(d) Manipulation of Lineage Balance Current Opinion in Genetics & Development

Conceptual diagram showing potential avenues for cell fate manipulation by designer transcription factors. (a) Direct activation of lineage specifier: expression of specific lineage specifier can be activated directly by targeting gene specific promoter or enhancer regions. The expression of lineage specifier later activates the downstream targets for differentiation; (b) Indirect activation of lineage specifier through network enhancer: Expression of specific lineage program could be activated by critical enhancer regions, which allows binding of multiple coregulator complexes and mediates downstream target expression through long range interaction; (c) Direct activation of lineage program through multiplexed activators: Multiple guide RNAs targeting the promoters of downstream lineage genes can be delivered and simultaneous activation of multiple targets in the lineage network can be achieved via the CRISPR effector system; (d) Manipulation of lineage balance: skewing of lineage differentiation can be achieved by concurrent activation of the preferred specifier and repression of the alternative specifier, thus tilting the balance. M: Mater regulators for specific cell lineage; PolII: RNA polymerase II transcription complex; P: Promoter. www.sciencedirect.com

Current Opinion in Genetics & Development 2014, 28:1–9

6 Cell reprogramming, regeneration and repair

Figure 3

PolII L

TF

L

T Transposon

Looping Linker

F

Locus Tracker

Directed Transposition

Physical Blocker

A/R

Activator/Repressor

E

Epigenetic modifier Current Opinion in Genetics & Development

A summary diagram showing the various applications of designer transcription factor in different technical settings. Different effector domains can be tethered to the DNA recognition domain of ZFPs and TALEs or the deactivated Cas9 protein. L: Looping factor domain; F: Fluorescent protein; A/R: Activation/Repression domain; TF: Transcription factor; T: Transposase domain; E: Demethylase/Histone modifier.

Acknowledgements This work is supported by Wellcome Trust (grant number 098051).

References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest 1.

Gurdon JB, Elsdale TR, Fischberg M: Sexually mature individuals of Xenopus laevis from the transplantation of single somatic nuclei. Nature 1958, 182:64-65.

2.

Tada M, Takahama Y, Abe K, Nakatsuji N, Tada T: Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr Biol 2001, 11:1553-1558.

3.

Davis RL, Weintraub H, Lassara B: Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 1987, 51:987-1000.

9.

Aoi T, Yae K, Nakagawa M, Ichisaka T, Okita K, Takahashi K, Chiba T, Yamanaka S: Generation of pluripotent stem cells from adult mouse liver and stomach cells. Science 2008, 321:699-702.

10. Liu H, Zhu F, Yong J, Zhang P, Hou P, Li H, Jiang W, Cai J, Liu M, Cui K et al.: Generation of induced pluripotent stem cells from adult rhesus monkey fibroblasts. Cell Stem Cell 2008, 3:587-590. 11. Liao J, Cui C, Chen S, Ren J, Chen J, Gao Y, Li H, Jia N, Cheng L, Xiao H et al.: Generation of induced pluripotent stem cell lines from adult rat cells. Cell Stem Cell 2009, 4:11-15. 12. Li W, Wei W, Zhu S, Zhu J, Shi Y, Lin T, Hao E, Hayek A, Deng H, Ding S: Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors. Cell Stem Cell 2009, 4:16-19. 13. Shu J, Wu C, Wu Y, Li Z, Shao S, Zhao W, Tang X, Yang H, Shen L, Zuo X et al.: Induction of pluripotency in mouse somatic cells with lineage specifiers. Cell 2013, 153:963-975.

4.

Xie H, Ye M, Feng R, Graf T: Stepwise reprogramming of B cells into macrophages. Cell 2004, 117:663-676.

14. Hou P, Li Y, Zhang X, Liu C, Guan J, Li H, Zhao T, Ye J, Yang W, Liu K et al.: Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 2013, 341:651-654.

5.

Takahashi K, Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126:663-676.

15. Niwa H, Miyazaki J, Smitha G: Quantitative expression of Oct-3/ 4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet 2000, 24:372-376.

6.

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.

7.

Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R et al.: Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318:1917-1920.

8.

Hanna J, Markoulaki S, Schorderet P, Carey BW, Beard C, Wernig M, Creyghton MP, Steine EJ, Cassady JP, Foreman R et al.: Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency. Cell 2008, 133:250-264.

Current Opinion in Genetics & Development 2014, 28:1–9

16. Thomson M, Liu SJ, Zou L-N, Smith Z, Meissner A, Ramanathan S: Pluripotency factors in embryonic stem cells regulate differentiation into germ layers. Cell 2011, 145:875-889. 17. Polo JM, Anderssen E, Walsh RM, Schwarz BA, Nefzger CM, Lim SM, Borkent M, Apostolou E, Alaei S, Cloutier J et al.: A molecular roadmap of reprogramming somatic cells into iPS cells. Cell 2012, 151:1617-1632. 18. Stadtfeld M, Maherali N, Breault DT, Hochedlinger K: Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell 2008, 2:230-240. www.sciencedirect.com

Creating the ‘‘Swiss Knives’’ for reprogramming Tsang et al.

19. Buganim Y, Faddah DA, Cheng AW, Itskovich E, Markoulaki S, Ganz K, Klemm SL, van Oudenaarden A, Jaenisch R: Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic phase. Cell 2012, 150:1209-1222. 20. Soufi A, Donahue G, Zaret KS: Facilitators and impediments of  the pluripotency reprogramming factors’ initial engagement with the genome. Cell 2012, 151:994-1004. This article examined the genome wide binding profile of OSKM at the early stage of reprogramming in human fibroblast. They identified large genomic regions that were refractory to OSKM binding. This study highlighted the limitation of reprogramming with native transcription factors. 21. Tiemann U, Sgodda M, Warlich E, Ballmaier M, Scho¨ler HR, Schambach A, Cantz T: Optimal reprogramming factor stoichiometry increases colony numbers and affects molecular characteristics of murine induced pluripotent stem cells. Cytometry A 2011, 79:426-435. 22. Papapetrou EP, Tomishima MJ, Chambers SM, Mica Y, Reed E, Menon J, Tabar V, Mo Q, Studer L, Sadelain M: Stoichiometric and temporal requirements of Oct4, Sox2, Klf4, and c-Myc expression for efficient human iPSC induction and differentiation. Proc Natl Acad Sci U S A 2009, 106:12759-12764. 23. Carey BW, Markoulaki S, Hanna JH, Faddah DA, Buganim Y, Kim J, Ganz K, Steine EJ, Cassady JP, Creyghton MP et al.: Reprogramming factor stoichiometry influences the epigenetic state and biological properties of induced pluripotent stem cells. Cell Stem Cell 2011, 9:588-598. 24. Stadtfeld M, Apostolou E, Akutsu H, Fukuda A, Follett P, Natesan S, Kono T, Shioda T, Hochedlinger K: Aberrant silencing of imprinted genes on chromosome 12qF1 in mouse induced pluripotent stem cells. Nature 2010, 465:175-181. 25. Theunissen TW, Costa Y, Radzisheuskaya A, van Oosten AL, Lavial F, Pain B, Castro LFC, Silva JCR: Reprogramming capacity of Nanog is functionally conserved in vertebrates and resides in a unique homeodomain. Development 2011, 138:4853-4865. 26. Okita K, Ichisaka T, Yamanaka S: Generation of germlinecompetent induced pluripotent stem cells. Nature 2007, 448:313-317. 27. Esch D, Vahokoski J, Groves MR, Pogenberg V, Cojocaru V, Vom Bruch H, Han D, Drexler HCA, Arau´zo-Bravo MJ, Ng CKL et al.: A unique Oct4 interface is crucial for reprogramming to pluripotency. Nat Cell Biol 2013, 15:295-301. 28. Jauch R, Aksoy I, Hutchins AP, Ng CKL, Tian XF, Chen J,  Palasingam P, Robson P, Stanton LW, Kolatkar PR: Conversion of Sox17 into a pluripotency reprogramming factor by reengineering its association with Oct4 on DNA. Stem Cells 2011, 29:940-951. This article demonstrated that a lineage specifier transcription factor SOX17 can be converted into a reprogramming factor by site specific enigneering of the coding sequence.

33. Pavletich NP, Pabo CO: Zinc finger-DNA recognition: crystal structure of a Zif268–DNA complex at 2.1 A˚. Science 1991, 252:809-817. 34. Sera T: Zinc-finger-based artificial transcription factors and their applications. Adv Drug Deliv Rev 2009, 61:513-526. 35. Barrow JJ, Masannat J, Bungert J: Neutralizing the function of a b-globin-associated cis-regulatory DNA element using an artificial zinc finger DNA-binding domain. Proc Natl Acad Sci U S A 2012, 109:17948-17953. 36. Bartsevich VV, Miller JC, Case CC, Pabo CO: Engineered zinc finger proteins for controlling stem cell fate. Stem Cells 2003, 21:632-637. 37. Ji Q, Fischer AL, Brown CR, Eastlund ER, Dvash T, Zhong B, Gerber MA, Lyons I, Knight SW, Kreader CA: Engineered zincfinger transcription factors activate OCT4 (POU5F1), SOX2, KLF4, c-MYC (MYC) and miR302/367. Nucleic Acids Res 2014, 4:1-10. 38. Maeder ML, Thibodeau-Beganny S, Osiak A, Wright Da, Anthony RM, Eichtinger M, Jiang T, Foley JE, Winfrey RJ, Townsend Ja et al.: Rapid ‘‘open-source’’ engineering of customized zinc-finger nucleases for highly efficient gene modification. Mol Cell 2008, 31:294-301. 39. Sander JD, Dahlborg EJ, Goodwin MJ, Cade L, Zhang F, Cifuentes D, Curtin SJ, Blackburn JS, Thibodeau-Beganny S, Qi Y et al.: Selection-free zinc-finger-nuclease engineering by contextdependent assembly (CoDA). Nat Methods 2011, 8:67-69. 40. Khalil AS, Lu TK, Bashor CJ, Ramirez CL, Pyenson NC, Joung JK,  Collins JJ: A synthetic biology framework for programming eukaryotic transcription functions. Cell 2012, 150:647-658. This article presented a comprehensive framework and the versatility of zinc finger desinger transcription factors in the study of transcription regulation. 41. Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S,  Lahaye T, Nickstadt A, Bonas U: Breaking the code of DNA binding specificity of TAL-type III effectors. Science 2009, 326:1509-1512. Both works [41,42] demonstrated the simple and elegant nucleotide recognition logic of TALEs. They laid the ground for construction of specific DNA binding TALE domain for designer transcription factor engineering. 42. Moscou MJ, Bogdanove AJ: A simple cipher governs DNA  recognition by TAL effectors. Science 2009, 326:1501. Both works [41,42] demonstrated the simple and elegant nucleotide recognition logic of TALEs. They laid the ground for construction of specific DNA binding TALE domain for designer transcription factor engineering. 43. Reyon D, Tsai SQ, Khayter C, Foden Ja, Sander JD, Joung JK: FLASH assembly of TALENs for high-throughput genome editing. Nat Biotechnol 2012, 30:460-465. 44. Schmid-Burgk JL, Schmidt T, Kaiser V, Ho¨ning K, Hornung V: A ligation-independent cloning technique for high-throughput assembly of transcription activator–like effector genes. Nat Biotechnol 2012, 31:76-81.

29. Wang Y, Chen J, Hu J-L, Wei X-X, Qin D, Gao J, Zhang L, Jiang J,  Li J-S, Liu J et al.: Reprogramming of mouse and human somatic cells by high-performance engineered factors. EMBO Rep 2011, 12:373-378. This article demonstrated that the reprogramming efficiency of native factor OSKM could be improved by modular engineering. A vrial transactviation domain VP16 was fused to individual OSKM to improve their gene activation ability.

45. Briggs AW, Rios X, Chari R, Yang L, Zhang F, Mali P, Church GM: Iterative capped assembly: rapid and scalable synthesis of repeat-module DNA such as TAL effectors from individual monomers. Nucleic Acids Res 2012, 40:e117.

30. Zhu G, Li Y, Zhu F, Wang T, Jin W, Mu W, Lin W, Tan W, Li W,  Street RC et al.: Coordination of engineered factors with TET1/ 2 promotes early-stage epigenetic modification during somatic cell reprogramming. Stem Cell Rep 2014, 2:253-261. This article demonstrated that modular fusion of the YAP transactivation domain with the native OSKM could improve their ability in recruiting demethylase TET1/TET2 in early stage of reprogramming.

47. Li L, Piatek MJ, Atef A, Piatek A, Wibowo A, Fang X, Sabir JSM, Zhu J-K, Mahfouz MM: Rapid and highly efficient construction of TALE-based transcriptional regulators and nucleases for genome modification. Plant Mol Biol 2012, 78:407-416.

31. Hirai H, Katoku-Kikyo N, Karian P, Firpo M, Kikyo N: Efficient iPS cell production with the MyoD transactivation domain in serum-free culture. PLoS ONE 2012, 7:e34149. 32. Rodrı´guez-Martı´nez JA, Peterson-Kaufman KJ, Ansari AZ: Smallmolecule regulators that mimic transcription factors. Biochim Biophys Acta 2010, 1799:768-774. www.sciencedirect.com

7

46. Sanjana NE, Cong L, Zhou Y, Cunniff MM, Feng G, Zhang F: A transcription activator-like effector toolbox for genome engineering. Nat Protoc 2012, 7:171-192.

48. Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P:  Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat Biotechnol 2011, 29:149-153. This work demonstrated that site-specific TALE designer transcription factor could be constructed by modular TALE repeat assembly to activate endogenous gene expression, including reprogramming factors such as Oct4 and Klf4 in imortalized cell line. Current Opinion in Genetics & Development 2014, 28:1–9

8 Cell reprogramming, regeneration and repair

49. Bultmann S, Morbitzer R, Schmidt CS, Thanisch K, Spada F, Elsaesser J, Lahaye T, Leonhardt H: Targeted transcriptional activation of silent oct4 pluripotency gene by combining designer TALEs and inhibition of epigenetic modifiers. Nucleic Acids Res 2012, 40:5368-5377. 50. Hu J, Lei Y, Wong W-K, Liu S, Lee K-C, He X, You W, Zhou R, Guo J-T, Chen X et al.: Direct activation of human and mouse Oct4 genes using engineered TALE and Cas9 transcription factors. Nucleic Acids Res 2014 http://dx.doi.org/10.1093/nar/ gku109. 51. Gao X, Yang J, Tsang JCH, Ooi J, Wu D, Liu P: Reprogramming to  pluripotency using designer TALE transcription factors targeting enhancers. Stem Cell Rep 2013, 1:183-197. This work demonstrated that TALE designer transcription factors could be used to study reprogramming. It also presented the genomic context of the targeting site could influence reprogramming. 52. Spitz F, Furlong EEM: Transcription factors: from enhancer binding to developmental control. Nat Rev Genet 2012, 13:613-626. 53. Smith E, Shilatifard A: Enhancer biology and enhanceropathies. Nat Struct Mol Biol 2014, 21:210-219. 54. Taberlay PC, Kelly TK, Liu C-C, You JS, De Carvalho DD, Miranda TB, Zhou XJ, Liang G, Jones PA: Polycomb-repressed genes have permissive enhancers that initiate reprogramming. Cell 2011, 147:1283-1294. 55. Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, Wong E, Orlov YL, Zhang W, Jiang J et al.: Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008, 133:1106-1117. 56. Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-Andre´ V, Sigova AA, Hoke HA, Young RA: Super-enhancers in the control of cell identity and disease. Cell 2013 http://dx.doi.org/10.1016/ j.cell.2013.09.053. 57. Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH, Rahl PB, Lee TI, Young RA: Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 2013, 153:307-319. 58. Crocker J, Stern DL: TALE-mediated modulation of transcriptional enhancers in vivo. Nat Methods 2013, 10:762-767. 59. Zhang Z, Xiang D, Heriyanto F, Gao Y, Qian Z, Wu W-S: Dissecting the roles of miR-302/367 cluster in cellular reprogramming using TALE-based repressor and TALEN. Stem Cell Rep 2013, 1:218-225. 60. Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, Zhang Y, Yang W, Gruber PJ, Epstein Ja et al.: Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 2011, 8:376-388. 61. Miyoshi N, Ishii H, Nagano H, Haraguchi N, Dewi DL, Kano Y, Nishikawa S, Tanemura M, Mimori K, Tanaka F et al.: Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell 2011, 8:633-638. 62. Konermann S, Brigham MD, Trevino AE, Hsu PD, Heidenreich M, Cong L, Platt RJ, Scott Da, Church GM, Zhang F: Optical control of mammalian endogenous transcription and epigenetic states. Nature 2013, 500:472-476.

66. Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE, Church GM: RNA-guided human genome engineering via Cas9. Science 2013, 339:823-826. 67. Cho SW, Kim S, Kim JM, Kim J-S: Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol 2013, 31:230-232. 68. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini La et al.: Multiplex genome engineering using CRISPR/Cas systems. Science 2013, 339:819-823. 69. Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J: RNAprogrammed genome editing in human cells. Elife 2013, 2:e00471. 70. Qi LS, Larson MH, Gilbert La, Doudna Ja, Weissman JS, Arkin AP,  Lim Wa: Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013, 152:1173-1183. This work demonstrated that the nuclease domain of the Cas9 protein could be deactivated by mutations and the deactivated Cas9 protein could be adapted to block mRNA transcription when directed to promoters. 71. Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, Torres SE, Stern Ginossar N, Brandman O, Whitehead EH, Doudna JA et al.: CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 2013, 154:442-451. This work demonstrated separate activation and repression domains could be tethered onto the deactivated Cas9 protein to regulate gene transcription. 72. Perez-pinera P, Kocak DD, Vockley CM, Adler AF, Kabadi AM, Polstein LR, Thakore PI, Glass KA, Ousterout DG, Leong KW et al.: RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat Methods 2013, 10:973-976. 73. Maeder ML, Linder SJ, Cascio VM, Fu Y, Ho QH, Joung JK: CRISPR RNA-guided activation of endogenous human genes. Nat Methods 2013, 10:977-979. 74. Cheng AW, Wang H, Yang H, Shi L, Katz Y, Theunissen TW, Rangarajan S, Shivalila CS, Dadon DB, Jaenisch R: Multiplexed activation of endogenous genes by CRISPR-on, an RNAguided transcriptional activator system. Cell Res 2013, 23:1163-1171. 75. Kearns NA, Genga RMJ, Enuameh MS, Garber M, Wolfe SA, Maehr R: Cas9 effector-mediated regulation of transcription and differentiation in human pluripotent stem cells. Development 2014, 141:219-223. 76. Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Dohmae N, Ishitani R, Zhang F, Nureki O: Crystal structure of cas9 in complex with guide RNA and target DNA. Cell 2014, 156:935-949. 77. Mak AN-S, Bradley P, Cernadas RA, Bogdanove AJ, Stoddard BL: The crystal structure of TAL effector PthXo1 bound to its DNA target. Science 2012, 335:716-719. 78. Deng D, Yan C, Pan X, Mahfouz M, Wang J, Zhu J-K, Shi Y, Yan N: Structural basis for sequence-specific recognition of DNA by TAL effectors. Science 2012, 335:720-723. 79. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agarwala V, Li Y, Fine EJ, Wu X, Shalem O et al.: DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 2013, 31:827-832.

63. Valton J, Dupuy A, Daboussi F, Thomas S, Mare´chal A, Macmaster R, Melliand K, Juillerat A, Duchateau P: Overcoming transcription activator-like effector (TALE) DNA binding domain sensitivity to cytosine methylation. J Biol Chem 2012, 287:38427-38432.

80. Pattanayak V, Lin S, Guilinger JP, Ma E, Doudna JA, Liu DR: Highthroughput profiling of off-target DNA cleavage reveals RNAprogrammed Cas9 nuclease specificity. Nat Biotechnol 2013, 31:839-843.

64. Deng D, Yin P, Yan C, Pan X, Gong X, Qi S, Xie T, Mahfouz M, Zhu J-K, Yan N et al.: Recognition of methylated DNA by TAL effectors. Cell Res 2012, 22:1502-1504.

81. Cho SW, Kim S, Kim Y, Kweon J, Kim HS, Bae S, Kim J-S: Analysis of off-target effects of CRISPR/Cas-derived RNAguided endonucleases and nickases. Genome Res 2014, 24:132-141.

65. Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, Romero DA, Horvath P: CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007, 315:1709-1712.

Current Opinion in Genetics & Development 2014, 28:1–9

82. Wu X, Scott DA, Kriz AJ, Chiu AC, Hsu PD, Dadon DB, Cheng AW, Trevino AE, Konermann S, Chen S et al.: Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat Biotechnol 2014 http://dx.doi.org/10.1038/nbt.2889. www.sciencedirect.com

Creating the ‘‘Swiss Knives’’ for reprogramming Tsang et al.

83. Li Y, Ehrhardt K, Zhang MQ, Bleris L: Assembly and validation of versatile transcription activator-like effector libraries. Sci Rep 2014, 4:4857. 84. Wang T, Wei JJ, Sabatini DM, Lander ES: Genetic screens in human cells using the CRISPR–Cas9 system. Science 2014, 343:80-84. 85. Koike-Yusa H, Li Y, Tan E-P, Velasco-Herrera MDC, Yusa K: Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library. Nat Biotechnol 2013, 32:267-273. 86. Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelsen TS, Heckl D, Ebert BL, Root DE, Doench JG et al.: Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 2014, 343:84-87. 87. De Wit E, Bouwman BAM, Zhu Y, Klous P, Splinter E, Verstegen MJAM, Krijger PHL, Festuccia N, Nora EP, Welling M et al.: The pluripotent genome in three dimensions is shaped around pluripotency factors. Nature 2013, 501:227-231.

www.sciencedirect.com

9

88. Apostolou E, Ferrari F, Walsh RM, Bar-Nur O, Stadtfeld M, Cheloufi S, Stuart HT, Polo JM, Ohsumi TK, Borowsky ML et al.: Genome-wide chromatin interactions of the Nanog locus in pluripotency, differentiation, and reprogramming. Cell Stem Cell 2013, 12:699-712. 89. Deng W, Lee J, Wang H, Miller J, Reik A, Gregory PD, Dean A, Blobel GA: Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell 2012, 149:1233-1244. 90. Chen B, Gilbert LA, Cimini BA, Schnitzbauer J, Zhang W, Li GW, Park J, Blackburn EH, Weissman JS, Qi LS et al.: Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 2013, 155:1479-1491. 91. Owens JB, Mauro D, Stoytchev I, Bhakta MS, Kim M-S, Segal DJ, Moisyadi S: Transcription activator like effector (TALE)directed piggyBac transposition in human cells. Nucleic Acids Res 2013, 41:9197-9207.

Current Opinion in Genetics & Development 2014, 28:1–9

Cellular reprogramming by transcription factor engineering.

Recent researches have identified multiple transcription factors as permissible reprogramming factors to pluripotency and lineage switching. The curre...
505KB Sizes 0 Downloads 4 Views