Skip to main content
Non-coding RNA Research logoLink to Non-coding RNA Research
. 2017 Apr 13;2(1):74–82. doi: 10.1016/j.ncrna.2017.04.002

The essentiality of non-coding RNAs in cell reprogramming

Joachim Luginbühl 1, Divya Mundackal Sivaraman 1, Jay W Shin 1,
PMCID: PMC6096403  PMID: 30159423

Abstract

In mammals, short (mi-) and long non-coding (lnc) RNAs are immensely abundant and they are proving to be more functional than ever before. Particularly in cell reprogramming, non-coding RNAs are essential to establish the pluripotent network and are indispensable to reprogram somatic cells to pluripotency. Through systematic screening and mechanistic studies, diverse functional features of both miRNA and lncRNAs have emerged as either scaffolds, inhibitors, or co-activators, necessary to orchestrate the intricacy of gene regulation. Furthermore, the collective characterizations of both miRNA and lncRNA reveal their interdependency (e.g. sequestering the function of the other) to modulate cell reprogramming. This review broadly explores the regulatory processes of cell reprogramming - with key functional examples in neuronal and cardiac differentiations - in the context of both short and long non-coding RNAs.

Keywords: miRNA, lncRNA, Reprogramming, Pluripotency, Neurons

1. Introduction

1.1. Regenerative medicine and reprogramming of cell state

Restoring normal functionality of diseased or injured tissues with healthy ones forms the crux of regenerative medicine [1]. During the embryonic development, cells terminally differentiate to more specialized cell fates with reduced cellular plasticity. However, numerous reprogramming methods have been developed to achieve pluripotency by reprogramming somatic cell to the pluripotent state or to another lineage [2]. This plasticity of the pluripotent stem cells to attain different cell fates provides a great potential to treat different diseases. Although the transfer of somatic cell nuclei [3] by oocytes [4] or sperm RNA [5] has shown the potential to reprogram cell states, transcription factor mediated reprogramming to generate induced pluripotent stem cells (iPSCs) [6] or mediate direct lineage conversion (trans-differentiation) [7] has gained considerable interest.

Introduction of exogenous transcription factors for somatic cell reprogramming leads to gross perturbations of the transcriptome and epigenome landscapes. This initiate a series of chromatin remodeling events that expose specific gene promoters and activate either pluripotency or cell type-specific gene regulatory networks (GRN) [8], [9]. More recently, reprogramming of iPSCs and direct lineage conversion was also achieved with small molecules instead of exogenous transcription factors [10]. Although considerable progress has been achieved in understanding the GRNs for reprogramming, low efficiency [11], genetic/epigenetic instability [12], [13], and in the case of direct lineage conversion, remnants of the initial cell fate, represent important issues that remain unsolved [14]. Moreover, several embryonic stem cell-specific cis-regulatory regions were not triggered during iPSC reprogramming [15]. Hence, there is a pressing need for identifying factors that can enhance reprogramming efficiency and maturation of converted cells, for example by taking cues from development [16] or through large-scale genetic screens [17], [18]. Notably, accumulating evidence suggests a central role of non-coding RNAs, an important class of regulators of gene expression and chromatin remodeling, during development and cell fate specification, which has prompted scientists to elucidate the potential role of non-coding RNAs for iPSC reprogramming and direct lineage conversion.

In the first part of this review, we will discuss micro-RNAs (miRNAs), a class of non-coding RNAs, which have been shown to – either in combination with the forced expression of specific sets of transcription factors or on their own – modulate the reprogramming of fully differentiated mature cells into iPSCs or transdifferentiation into cells of a different lineage while bypassing an intermediate pluripotent state. In the second part, we will summarize the role of another class of non-coding RNAs, long non-coding RNAs (lncRNAs), in cell reprogramming. We rationalize that miRNAs and lncRNAs represent a promising and powerful tool for the future of reprogramming and regenerative medicine.

2. miRNAs: master orchestrators of gene expression

Since the discovery of the very first micro-(mi) RNA, lin-4, in Caenorhabditis elegans (C.elegans) [19], [20], thousands of micro-(mi) RNAs have been identified in both, plants and animals, where they have the capacity to interact with RNA, DNA and protein molecules to regulate gene expression at multiple levels. The biogenesis and function of miRNAs have been comprehensively reviewed elsewhere [21], [22]. Briefly, miRNAs are first transcribed by RNA polymerase II/III into long, primary transcripts called pri-miRNAs. Thereafter, pri-miRNAs are cut and processed into short pri-miRNAs of approximately 70–100 nucleotides in length which fold into a hairpin shape. This is mediated by a microprocessor complex consisting of a ribonuclease III protein called Drosha, a RNA-binding protein called DGCR3 and several other cofactors [23], [24], [25], [26]. After export into the cytoplasm, either a RNase III called Dicer or an Argonaute protein, Ago2, converts pri-miRNAs into short, double-stranded miRNA molecules of approximately 22 nucleotide length [27], [28]. One of the strands (guide-strand), which is complementary to its target, then is recruited by argonaute protein to form the miRNA-induced silencing complex (miRISC), with which the miRNA is usually guided to the 3′ UTR of its target mRNA. The other strand (passenger-strand), which is not complementary to target mRNA, has long been thought to be degraded during miRNA biogenesis [29], but there is mounting evidence that passenger strand miRNAs can also target mRNAs and exert functions for example during cancer formation [30], [31]. However, in order to regulate gene expression, miRNAs not only bind to RNA, but they also have the potential to interact with DNA and protein molecules, which attributes them as master regulators in orchestrating GRNs [32], [33], [34], [35], [36], [37], [38], [39].

2.1. The role of miRNAs in iPSC reprogramming

Gene regulatory networks (GRNs) that instruct developmental fates of cells are typically composed of specific sets of transcription factors, epigenetic modulators, signaling molecules and non-coding RNAs [40], [41], [42], [43]. Advances in the field of cell reprogramming over the last decades have substantiated the importance of manipulating these GRNs in order to render different cell fates interchangeably [44], [45], [46], [47]. Given the key role of miRNA in modulating GRNs, it is not surprising that manipulation of a variety of miRNAs has emerged as a potent method to either enhance or inhibit the reprogramming towards diverse cell types. Moreover, there is a growing number of miRNAs that have been shown to, on their own, have the capacity to reprogram cell fates. These findings underscore the importance of realizing the various functions of miRNAs in modulating GRNs, which will undoubtedly lead to the discovery of many more miRNAs with critical roles in cell reprogramming and cell fate decisions in coming years.

A common strategy for the identification of candidate miRNAs involved in iPSC reprogramming has been to analyze differently expressed miRNAs (profiled by means of microarrays or small RNA-sequencing) between pluripotent stem cells and differentiated cells during iPSC generation or during the differentiation into somatic cells [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59]. An example of miRNAs that are downregulated during differentiation of ESCs and improve iPSC formation is a subset of the miR-290 cluster called the ES cell-specific cell cycle-regulating (ESCC) miRNAs, which includes miR-291-3p, miR-294 and miR-295 [60]. Introduction of miR-291-3p, miR-294 and miR-295 along with Oct-4, Sox2 and Klf4 (OSK) into mouse embryonic fibroblasts (MEFs) was found to consistently increase the number of Oct4-positive iPSC colonies compared with MEFs transduced with OSK alone [49]. Strongest improvement of iPSC formation was observed after introduction of miR-290, while other members of the same cluster, miR-292-3p and miR-293, had no effect. Interestingly, c-Myc binds to the promoter of these miRNAs, suggesting that they act downstream of c-Myc. In another study, mimics of human miR-302b and miR-372, which are orthologous to the mouse miR-291, miR-294 and miR-295, were able to enhance iPSC reprogramming efficiency [61]. While the subset of the miR-290 cluster described above was selected based on its expression during ESC differentiation, another study selected candidate miRNAs based on their upregulation during the early stages of iPSC reprogramming [51]. Overexpression of two members of the miR-106a cluster, miR-93 and miR-106b greatly enhanced iPSC generation, while knockdown of the same miRNAs as well as another member of the same cluster, miR-25, using miR-inhibitors decreased reprogramming efficiency. Further analyses revealed that both, miR-93 and miR-106a repressed TGFBR2 and p21 expression. Paradoxically, inhibiting TGFBRI kinases undermined ES cell renewal, whereas small molecule inhibitors of TGFBR1 enhanced iPSC reprogramming. Thus, factors that are essential for ESC self-renewal do not necessarily improve iPSC formation, but might even represent barriers for cell reprogramming.

Temporal gene expression profiling revealed that iPSC generation from MEF involves a multistep process characterized by initiation, maturation and stabilization phases [56]. In-depth analysis of the dynamic gene expression in combination with a systematic genetic RNAi screen revealed that the initiation phase is characterized by a coordinated mesenchymal-to-epithelial transition and BMP signaling, as well as upregulation of several miRNAs [56]. Inhibition of BMP signaling during the initiation phase suppressed the expression of multiple miRNAs including miR-200a, −200b, and −205, whereas exogenously supplied BMP enhanced their expression. Importantly, transfection of MEF with two miR-200 family mimics, Mim-200b and Mim-200c, in conjunction with OSKM, stimulated MET and accelerated through the initiation phase. Mechanistic insight into the synergism of members of the miR-200 cluster and OSKM during iPSC reprogramming revealed that Oct4 and Sox2 directly target the promoters of members of the miR-200 cluster and activate their expression [57]. Upon activation, miR-200 family members mainly targeted ZEB2 through directly binding to its 3′ UTR, thereby promoting mesenchymal-to-epithelial transition and enhancing iPSC generation.

In contrast to ESCC miRNAs, which are highly enriched in mouse ESCs (mESCs), miR-21 and miR-29a belong to the most abundant miRNAs in MEF [59]. Inhibition of their expression using miRNA inhibitors together with overexpression of the OSKM factors has been shown to enhance iPSC reprogramming efficiency [59]. c-Myc was found to play a predominant role in suppressing miR-21 and miR-29a at the transcriptional level. Analysis of the mechanism by which miR-21 and miR-29a affect reprogramming showed that both miRNAs inhibit expression of p53, which has a well-known role in modulating iPSC reprogramming [62], [63], [64], [65]. Indeed, the p53 pathway, widely known for its role in tumor suppression, has recently emerged as a central roadblock for iPSC generation (reviewed in Ref. [66]). Although most of the targets of p53 are protein-coding genes, several miRNAs are vital components of the p53 pathway, which has prompted researchers to investigate their role during cell reprogramming. miR-34, miR-145 and miR199a, all of which are induced by p53, have been shown to inhibit iPSC generation via different mechanisms [67], [68], [58], [69] (Fig. 1). Conversely, miR-138 has been shown to directly target p53, and several miRNAs, including miR-93, miR-106a/b and miR290, target p21, a well-known component of the p53 pathway, thereby promoting iPSC generation [70], [51], [71]. Together with miR-21 and miR-29a, these miRNAs therefore constitute important modulators of the p53 GRN, locating it at the crossroads between cell reprogramming and tumor suppression.

Fig. 1.

Fig. 1

Non-coding RNA-centered Gene Regulatory Network (GRN) in cell reprogramming. (A) A simplified schematic summarizing miRNAs and lncRNAs with known roles in modulating GRNs involved in cell reprogramming downstream of p53 and PTB. (B + C) The lncRNAs linc-RoR (B) and GAS5 (C) act as competitive inhibitors termed “miRNA sponges”, containing multiple tandem binding sites for miRNAs which inhibit cell reprogramming. (D) During development, miR-124 promotes neuronal differentiation by targeting REST, while being inhibited by REST in a feedback loop. The RNA-binding protein PTB suppresses targeting of REST by multiple miRNAs including miR-124. REST and PTB ultimately converge in repressing many neuronal genes, and either overexpression of miR-124 or downregulation of PTB enhances direct reprogramming of fibroblasts into neurons.

Interestingly in a more recent study, miR-29a has also been reported to target DNA demethylases [72]. Repression of miR-29a led to the demethylation of fibroblast-specific DNA marks, and iPSC lines derived from miR-29a repression reduced the aberrant DNA methylation status observed in control-derived iPSCs and resembled human ESCs (hESCs) in their methylome. Conversely, another member of the miR-29 cluster, miR-29b, is upregulated by SOX2 and targets DNA methyltransferases (Dnmt3a and Dnmt3b), thereby enhancing iPSC formation [73]. Thus, two members of the same miRNA cluster, which both target epigenetic modulators, can have opposite roles during iPSC reprogramming. Notably, recent studies indicate that miR-29c, the third member of the miR-29 cluster, also targets Dnmt3a and Dnmt3b, indicating that miR-29c might also have a role during the formation of iPSCs, but this remains to be tested [74], [75], [76].

An alternative approach for the identification of candidate miRNAs is to perform high-throughput genetic screens [17], [18]. Using MEFs carrying an Oct4-GFP reporter gene – which allows to measure the efficiency of iPSC generation by quantifying GFP expression – screened 570 chemically synthesized mouse miRNA mimics for their effect on iPSC reprogramming [17]. While validating previously known miRNAs, this screen identified miR-181, which was previously not associated with reprogramming, to promote iPSC generation. Notably, neither MEFs nor pluripotent stem cells express high levels of miR-181. Instead, miR-181 expression is transiently induced during the initial phase of iPSC reprogramming and subsequently silenced in iPSCs. A siRNA screen for targets of miR-181 and miR-294, an ESCC miRNA, furthermore revealed that during reprogramming initiation, the miR-181 and ESCC miRNA families have different targets, but come together when inhibiting TGF-β signaling and activating Wnt signaling, while ESCC miRNAs alone activated Akt signaling. In another screening study, several new miRNAs including miR130, miR-301 and miR-721 were found to enhance reprogramming of fibroblasts into iPSCs [18]. Analysis of target genes revealed that all three miRNA downregulated protein expression of the transcription factor Meox2 at the translational level. Transient silencing of Meox2 in MEFs significantly increased iPSC colony formation, albeit at lower levels than overexpression of the single miRNA, indicating that additional downstream effectors of these miRNA could have beneficial effects on iPSC reprogramming.

There are reports of miRNA that are able to reprogram iPSCs without additional pluripotency-associated transcription factors. For example, the miR302/367 cluster, which is highly expressed in ESCs and iPSCs, is composed of five miRNAs, four of which –miR302a/b/c/d-share an identical seed sequence [77]. The fifth miRNA, miR-367, has a different seed sequence and therefore is likely to target different mRNA transcripts. Lentiviral overexpression of miR302a/b/c/d was able to efficiently reprogram MEF and human fibroblasts to a pluripotent state in the absence of any additional pluripotent stem cell transcription factor [78]. Moreover, reprogramming using these miRNAs alone was significantly more efficient than with the previously identified OSKM factors. In the absence of miR-367, Oct4 expression was not induced, revealing that miR-367 activates Oct4 expression during reprogramming. Interestingly, in addition to the expression of miR-367, the HDAC inhibitor valproic acid (VPA) was essential for iPSC formation during reprogramming of MEF, while VPA was not required for the generation of iPSCs from human fibroblasts. VPA specifically degrades the HDAC2 protein, which is expressed at higher levels in MEF than in human fibroblasts. Thus, this study revealed that low levels of HDAC2 is critical during iPSC reprogramming, and that overexpression of miRNAs of the miR302/367 cluster without additional transcription factors required activation of Oct4 expression as well as epigenetic modulators. Similarly, combined transfection of miR200c, miR-302s and miR-369 was shown to be able to reprogram mouse and human somatic cells to pluripotency without additional transcription factors, albeit at very low efficiency (0.03%) [53]. Transfection of only one or two members in any combination did not generate iPSC colonies. Bisulfite sequencing revealed that CpG dinucleotides of the promoters of Nanog and Oct4 at later stages during reprogramming were less frequently methylated, indicating reactivation of immature status-related genes. Furthermore, in a recent study, knockout of miR-369 inhibited OSKM-mediated reprogramming of adipose-derived mesenchymal stem cells into iPSCs [79]. Overexpression of miR-369 was found to stabilize splicing factors of pyruvate kinase, which led to increased iPSC generation. If optimized further, the miRNA-only strategy will be of great interest for a clinical application, as it does not employ any integrating viral vectors.

2.2. miRNAs in transdifferentiation

Transdifferentiation, or direct cell reprogramming, refers to the direct conversion of a fully differentiated cell into another without passing through a pluripotent stage [80]. In the field of regenerative medicine, direct cell reprogramming is of particular interest due to the decreased risk of tumor formation and the potential to generate more homogenous cell populations with regard to reproducibility and subtype generation [81]. Neurons and cardiomyocytes are among the pivotal therapeutically relevant cell types, and several studies have addressed the possibility to directly convert differentiated cells into neurons or cardiomyocytes using either transcription factors, miRNAs, small molecules or a combination thereof (reviewed in Refs. [82], [83]). While direct reprogramming of neurons and cardiomyocytes have been extensively discussed elsewhere [84], [85], [82], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [80], [98], [81], [83], we will focus on studies that revealed different miRNAs with a role in mediating direct cell reprogramming towards neurons and cardiomyocytes.

Post-mitotic neural development and dendrite morphogenesis are mediated by repression of BAF53a by miR-9 and miR-124 [99]. Lentiviral overexpression of miR-9/9–124 along with the neurogenic factor NeuroD2 directly converted human fibroblasts into post-mitotic neurons expressing mature neuronal markers such as Map2 and firing action potentials [100]. Addition of two other neurogenic factors, Ascl1 and Myt1l, further increased maturation of the generated neurons and enhanced efficiency of direct conversion. During the process of neuronal differentiation, all subunits of the nBAF complex were upregulated (Baf53b, 45b and 45c), while miR-9/9–124 repressed Baf53a. Thus, miR-9/9–124 act on multiple targets during the reprogramming of human fibroblasts into induced neurons. In another study, combination of overexpression of miR-9/9–124 with transcription factors enriched in the developing striatum – BCL11B, DLX1, DLX2 and MYT1L-led to the direct conversion of human postnatal and adult fibroblasts into enriched population of striatal medium spiny neurons (MSN) [101]. Transfection of the striatal transcription factors without miR-9/9–124 was ineffective in generating neurons. After transplantation into the mouse brain, the induced MSN survived for up to 6 months and established typical connections to the anatomical targets of MSN. Indeed, miR-124 seems to take an important role in both, regulating normal neuronal differentiation as well as promoting conversion of various cell types into cells of the neuronal lineage. For instance, miR-124 regulates the transcription silencing complex built on RE1-silencing transcription factor (REST), which in turn represses many neuronal-specific genes in non-neuronal cells, including miR-124 itself [102], [103]. In a seminal study, suppression of polypyrimidine-tract-binding (PTB) protein, a target of miR-124, and its homolog nPTB in different cell types including HeLa cells, human embryonic carcinoma stem cells, mouse neuronal progenitor cells, human retinal epithelial cells and MEFs led to the direct conversion into functional neurons [104]. Subsequent studies revealed that PTB downregulation dismantles multiple components of the REST complex through different mechanisms, including direct competition with miRNA-targeting or boosting miRNA action on specific genes by changing local RNA secondary structure, thereby shielding or enhancing miRNA target sites in adjacent regions. Since components of the REST complex show several polypyrimidine-tract-binding (PTB) protein binding sites which are previously validated target sites of miR-124, miR-9 and miR-449 [105], [106], these results suggest that PTB downregulation enables dismantling of the REST complex by multiple components of the miRNA machinery. As a result, critical REST-repressed transcription factors such as Ascl1, Brn2, Zic1, Olig2, and NeuroD1 – previously shown to induce transdifferentiation of MEFs into neurons [107] – were upregulated.

In another example of miRNAs' involvement in transdifferentiation, transient transfection of mouse fibroblasts with miR-1, miR-33, miR-208 and miR-499 (miR-combo) was able to convert them into functional cardiomyocytes [108]. Addition of the small molecule JAK inhibitor I drastically enhanced this process, while other compounds tested had no beneficial effect. Furthermore, intramyocardial injection of the same miRNAs immediately after injury by permanent ligation of the left ascending coronary artery revealed conversion of cardiac fibroblasts into cardiomyocytes in vivo. Notably, the same group identified a specific media composition consisting of advanced DMEM/F12, ascorbic acid and insulin-transferrin-selenium, which increased conversion efficiency of the miR-combo by up to 15 fold [109]. More recently, another study revealed that addition of miR133a to the transcription factors Gata4, Mef2c and Tbx5 improved cardiac reprogramming of mouse and human fibroblasts through down-regulation of Snai1, a master regulator of epithelial to mesenchymal transition [110]. Inclusion of miRNA133a significantly increased the number of beating induced cardiomyocyte-like cells and shortened the duration until the appearance of beating cells.

However, miRNAs have also proven to be essential for the direct reprogramming of other cell types other than neurons and cardiomyocytes. Transfection of astrocytes with viral particles expressing miR-302/367 has been shown to directly convert human astrocytes into neuroblasts [111]. This conversion required addition of VPA. Importantly, during a 2 month follow up study, no tumor formation of the converted neuroblasts was observed, making this approach promising for the treatment of various neurological diseases. In a different study, overexpression of miR302/367 together with miR9/9–124 in human fibroblasts led to the direct conversion into neurons [112]. The converted neurons upregulated synaptic markers in vitro and in vivo, indicating establishment of connectivity. Interestingly, several miRNAs including miR-302 have also been shown to facilitate conversion of pancreas progenitor cells. Co-transfection of miR-302 with PDX1, NGN3 and MAFA into a human hepatic cell line activated expression of pancreas progenitor cell-related gene expression, leading to the generation of insulin and C-peptide producing cells when combined with a chemically defined medium [113]. Thus, miR-302 may represent a more general miRNA facilitating the indirect reprogramming of various cell types into pluripotent cells or the direct conversion into different somatic cells.

Taken together, research over the last years has enlightened the great potential of miRNAs to regulate several gene regulatory networks involved in cell reprogramming. Further elucidation of the diverse functions of miRNAs during iPSC reprogramming and transdifferentiation will not only enhance our understanding of the underlying mechanisms, but also accelerate the establishment of cell reprogramming in regenerative medicine.

3. Long non-coding RNAs

The Encyclopedia of DNA Elements (ENCODE) and the Functional Annotation of the Mammalian Genome (FANTOM) projects have shown that the majority of products of the mammalian genome are long non-coding RNAs (lncRNAs) with yet uncharacterized functions [114], [115], [116]. Usually defined as non-protein coding transcripts longer than 200 nucleotides, lncRNAs are transcribed by RNA polymerase II and further capped and spliced, similar to coding mRNA. Hence, from a biochemical point of view, lncRNAs are not distinct from mRNA. However, lncRNAs lack an open reading frame (ORF) and a large proportion of them are not poly-adenylated. Until now, lncRNAs have been shown to function by modulating the transcriptome via the regulation of epigenetic, transcriptional, and post-transcriptional mechanisms [117]. lncRNAs acts as (i) signals, mirroring transcriptional processes and hence may represent markers of biological events; (ii) decoys for miRNA target sites which titrate away miRNAs and regulatory proteins; (iii) guides that recruit chromatin remodeling factors/enzymes in cis or trans; and (iv) scaffolds, enabling the formation of ribonuclear protein complexes involved in gene regulatory events [118], [119]. Thus, the intracellular localization of lncRNAs within chromatin, sub-nuclear domains, the nucleoplasm or the cytoplasm can provide vital clues to their mechanism of action [120]. Furthermore, lncRNAs have been shown to be poorly conserved when compared to other small non-coding RNAs, suggesting that the secondary structure may represent their main functional unit [121].

3.1. LncRNAs as additional regulatory layer in reprogramming and pluripotency

Pluripotency-associated networks of genes, including the core transcription factors and aforementioned miRNAs, govern the pluripotent state [45]. Precise understanding of this regulatory network is essential for somatic cell reprogramming so as to modulate the epigenetic state of somatic cell towards pluripotency. Recent evidence indicates that lncRNAs function as an additional regulatory layer in maintaining pluripotency and determining cell fate [122], [123], [124], [125], [126], [127], [128], [129], [45], [130], [131]. Thus, lncRNAs may form additional nodes in the gene regulatory network (GRN), either as upstream elements for maintaining pluripotency/cell fate conversion or as alternative paths for reprogramming. Delineating the regulatory role of lncRNAs in core GRNs controlling the acquisition of pluripotency or mediating cell fate conversion requires comprehensive understanding of lncRNA expression profiles during the conversion of pluripotent cells to a defined fate or during reprogramming of somatic cells into iPSCs. In this section, we will focus on the role of lncRNAs in reprogramming, and subsequently provide examples of lncRNAs that have been shown to modulate cell fate conversion to the neural lineage.

3.2. LncRNAs and reprogramming

Lineage-committed cells are characterized by distinct regulatory epigenetic marks that safeguard cellular identity [132]. Therefore, derailing this epigenetic barrier is the prime event initiating cell fate conversion. Forced transformations of cell fates, by manipulating expression of pluripotency/lineage-related transcription factors, results in drastic epigenetic modifications coupled with widespread transcriptomic changes [16], [9]. The regulatory potential of lncRNAs in governing epigenetic and transcriptomic changes is well established, and growing evidence attributes lncRNAs as an important regulator during cell reprogramming.

While characterizing the transcriptional dynamics during iPSC formation, a study by Loewer et al. revealed 10 lncRNAs that are involved in achieving pluripotency [133]. Among them, regulator of reprogramming lncRNA (lincRNA-RoR) emerged as an important candidate whose perturbation resulted in altered reprogramming efficiency of fibroblasts to iPSCs [133]. The mechanistic role of lincRNA-RoR was subsequently characterized, where it acts as a key competing endogenous RNA for the network of miRNAs regulating core transcription factors involved in pluripotency. Most importantly, lincRNA-RoR shares seed sequences with miR-145, the miRNA that represses expression of core pluripotency transcription factors [134], [69]. iPSCs express lincRNA-RoR at a higher level than miR-145, thus enabling it to shield miR-145 target sites and preventing miRNA-mediated repression.

Later, a large intergenic noncoding RNA p21 (lincRNA-p21) was identified, which impaired reprogramming by sustaining H3K9me3 and CpG methylation at pluripotency gene promoters [135]. While, lincRNA-p21 is induced by p53 during reprogramming, it does not promote apoptosis or cellular senescence, but interacts with methyltransferases SETDB1 and DNMT1 through the RNA-binding protein HNRNPK to maintain the repressive chromatin state [135]. LncPRESS is another p53-induced lncRNA crucial for maintenance of ESC identity. This particular lncRNA physically interacts with the histone deacetylase SIRT6 and restricts its chromatin localization, resulting in high levels of histone H3K56 and H3K9 acetylation at pluripotency gene promoters. Depletion of lncPRESS1 induced lineage-specific genes with concurrent reduction in expression of pluripotency genes [136].

Once pluripotency is achieved, lncRNAs also regulate proliferation and self-renewal of iPSCs. Initial studies have attributed lncRNAs in the maintenance of pluripotency based on their co-expression with pluripotency related genes [137]. In hESCs, a pioneering study revealed three lncRNAs (AK056826, EF565083, BC026300) that are exclusively expressed in undifferentiated hESCs/iPSCs and co-expressed with key pluripotency drivers [138], and depletion of these lncRNAs promoted differentiation.

Later, systematic loss-of-function experiments identified several lncRNAs that regulate core pluripotency transcription factors. Utilizing a genome-wide shRNA screen of mESC transcripts together with chromatin immunoprecipitation of key pluripotency transcription factors, the lncRNAs Rncr2/AK028326 (Oct4-activated) and AK141205 (Nanog-repressed) were identified to maintain pluripotency [139]. Rncr2/AK028326 furthermore was shown to act as a co-activator of Oct4 in a regulatory feedback loop [139]. Another genome-wide siRNA/shRNA screen in mESCs revealed additional 26 lncRNAs regulating the pluripotent state, which was validated using a Nanog luciferase reporter system [140], and Panct1, Panct2, Panct3 (AK081885) to regulate mESC identity [141]. Panct1 was followed up for depletion studies, which led to downregulation of pluripotency related gene expression, but this study lacked mechanistic explanation for Panct1 function. A more recent large scale ‘loss of pluripotency’ screen, which employed CRISPR interference (CRISPRi) targeting 16,401 lncRNAs in iPSCs, indicated that inhibition of only 9 lncRNA loci reduced POU5F1/OCT4 expression [142].

Multiple studies revealed that lncRNAs regulate pluripotency by a variety of mechanisms including histone modifications, as scaffolds and by forming competing endogenous RNA (ce-RNA) for miRNAs that repress expression of pluripotency genes. Apart from the evidences of co-expression with pluripotency-related genes, Dinger et al. identified developmentally regulated lncRNAs in mESCs named Evx1as and Hoxb5/6as that physically interacted with tri-methylated H3K4 histones and histone methyltransferase MLL1, suggesting epigenetic mechanisms that coordinate pluripotency and differentiation [137]. Notably, lncRNA also regulate H3K27 methylation at promoters of differentiation-related genes, resulting in their transcriptional repression. H3K27 di/trimethylation involves the polycomb repressive complex 2 (PRC2), and the ARID domain-containing protein (JARID2) forms an essential accessory factor for the proper recruitment of PRC2 [143]. Several lncRNA encoded within the imprinted Dlk1-Dio3 locus were reported to bind to JARID2. Of these, MEG3 binding promoted recruitment and assembly of PRC2 at differentiation-related genes - maintaining the pluripotency of hiPSCs [144].

A recent study revealed that transition from pluripotency to differentiation required downregulation of ribosomal RNA synthesis in the nucleolus, resulting in ribosomal DNA heterochromatinisation [145]. The lncRNA pRNA was shown to interact with nucleolar repressor factor TIP5 to promote both H3K27 and H3K9 tri-methylation during differentiation of mESCs. Thus, depletion of pRNA exemplified a role of lncRNA modulating pluripotency through regulation of chromatin remodeling, retaining ESCs in a pluripotent state exhibiting a reduced number of heterochromatic loci.

LncRNAs can also function as scaffolds to maintain pluripotency by promoting recruitment of transcription factors and other chromatin modifiers [136]. Recently, human endogenous retrovirus subfamily H (HERVH) transcripts were identified to function as lncRNAs, forming a scaffold which recruits p300 and OCT4 to HERVH LTR7 regions to regulate pluripotency-associated transcripts [146]. Additionally, Tcl1 Upstream Neuron-Associated lincRNA (TUNA) functions as a scaffolding-lncRNA, forming a complex with RNA-binding proteins that binds to the promoters of pluripotency drivers [147]. Recently, a lncRNA - AK048794 was also shown to act as competing endogenous ce-RNA for miR-592, resulting in de-repression of its endogenous target, FAM91A1, which is involved in maintenance of pluripotency [148]. Likewise, growth-arrest-specific transcript 5 (GAS5), which is highly expressed in hESCs and whose expression correlated with hESC self-renewal, functions as ce-RNA, reducing targeting of miRNA to the TGF beta receptor family ligand NODAL, thereby sustaining NODAL expression and promoting hESC self-renewal [149]. Taken together, these reports revealed a complex regulatory network of lncRNAs that orchestrates acquisition and maintenance of pluripotency.

3.3. LncRNAs involved in neural fate decision: evidences from ESCs and iPSCs

Embryonic neural development is characterized by the expression of numerous lncRNAs whose role in promoting neural differentiation has been well characterized [150]. Although the large number of spatio-temporally expressed lncRNAs underscores their functionality, the underlying regulatory mechanisms of many of them remain elusive, demanding for large-scale perturbation studies to characterize their function [142].

In one comprehensive study pertaining to neural fate determination of hESCs, Ng et al. identified a group of 35 lncRNAs involved in differentiation of hESCs into neurons. Mechanistic studies revealed that these lncRNAs physically interacted with the nuclear proteins REST and SUZ12 [138]. In a subsequent study, one of these lncRNAs, rhabdomyosarcoma 2-associated transcript (RMST), was shown to form an indispensable regulatory complex required for SOX2 binding to the promoter regions of neurogenic transcription factors, thereby co-regulating downstream genes implicated in neurogenesis [151]. In another study, depletion of TUNA resulted in transition from pluripotency towards a neural cell fate [147]. Mechanistically, TUNA formed a complex with three RNA-binding proteins, PTBP1, hnRNP-K, and Nucleolin, regulating key pluripotency-associated genes Nanog, Sox2, and Fgf4 [147]. In another example, HOTAIRM1, a cis-acting regulator of the HOXA gene cluster and CRNDE, located in proximity to the IRX5 gene which is involved in brain patterning and neurogenesis, were upregulated in human iPSCs that differentiated into neurons [152]. Furthermore, a recent report revealed that the loss of non-coding RNA expression form the DLK1-DIO3-imprinted locus correlated with reduced neural differentiation potential of hESC lines [153]. Together, these studies revealed a large number of lncRNAs that are differentially expressed during the differentiation of hiPSCs to neurons, but comprehensive and systematic characterization of their function is still lacking.

Notably, lncRNAs that regulate direct cell reprogramming have not been identified yet. The abundance of lncRNAs in the mammalian genome demands for large-scale perturbation studies during direct cell programming coupled with molecular phenotyping and functional characterization. Given the prominent role of lncRNAs in epigenetic control and the fact that transdifferentiation involves epigenetic modifications, such studies are likely to reveal lncRNAs involved in direct reprogramming of different cell types including neurons [125], [154].

4. Future perspectives

Discovery of the increasingly diverse characteristics of miRNA and lncRNAs requires a deeper mechanistic exploration into localization, structure, and interacting partners to decipher their involvement in cell reprogramming. Additional studies that use large-scale transcript perturbation techniques such as CRISPR/Cas9 will provide an unprecedented advantage for functionally characterizing novel non-coding RNAs [142]. Moreover, single-cell transcriptome analysis in ESCs/IPSCs have been employed to reveal dynamically expressed miRNA and lncRNAs at different stages of reprogramming [155], [156]. With rapid reduction in cost for sequencing, the single-cell resolution to study gene expression profiling will allow highly accurate and efficient isolation of target cell types, making it suitable technology for both basic and clinical stem cell research. As more mechanisms of non-coding RNAs are revealed, additional opportunities to devise techniques to generate high quality iPSCs and directly converted cell types will become prevalent – accelerating the application of reprogrammed cells in the clinics.

Declaration of interest

Authors claim no conflict of interest.

Acknowledgements

This work was supported by Research Grant from MEXT to the RIKEN Center for Life Science Technologies and Grant-in-aid for Scientific research for JSPS fellow (JL).

Contributor Information

Joachim Luginbühl, Email: joachim.luginbuhl@riken.jp.

Divya Mundackal Sivaraman, Email: divya.sivaraman@riken.jp.

Jay W. Shin, Email: jay.shin@riken.jp.

References

  • 1.Mao A.S., Mooney D.J. Regenerative medicine: current therapies and future directions. Proc. Natl. Acad. Sci. 2015;112:14452–14459. doi: 10.1073/pnas.1508520112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kwon D., Ji M., Lee S., Seo K.W., Kang K.S. Reprogramming enhancers in somatic cell nuclear transfer, iPSC technology, and direct conversion. Stem Cell Rev. 2017;13(1):24–34. doi: 10.1007/s12015-016-9697-x. [DOI] [PubMed] [Google Scholar]
  • 3.Gurdon J.B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. Development. 1962;10:622–640. [PubMed] [Google Scholar]
  • 4.Jullien J., Pasque V., Halley-Stott R.P., Miyamoto K., Gurdon J.B. Mechanisms of nuclear reprogramming by eggs and oocytes: a deterministic process? Nat. Rev. Mol. Cell Biol. 2011;12:453–459. doi: 10.1038/nrm3140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Chen Q., Yan W., Duan E. Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications. Nat. Rev. Genet. 2016;17:733–743. doi: 10.1038/nrg.2016.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Takahashi K., Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 7.Davis R.L., Weintraub H., Lassar A.B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 1987;51:987–1000. doi: 10.1016/0092-8674(87)90585-x. [DOI] [PubMed] [Google Scholar]
  • 8.Krause M.N., Sancho-Martinez I., Izpisua Belmonte J.C. Understanding the molecular mechanisms of reprogramming. Biochem. Biophys. Res. Commun. 2016;473:693–697. doi: 10.1016/j.bbrc.2015.11.120. [DOI] [PubMed] [Google Scholar]
  • 9.Xu Y., Zhang M., Li W., Zhu X., Bao X., Qin B., Hutchins A.P., Esteban M.A. Transcriptional control of somatic cell reprogramming. Trends Cell Biol. 2016;26:272–288. doi: 10.1016/j.tcb.2015.12.003. [DOI] [PubMed] [Google Scholar]
  • 10.Lin T., Wu S. Reprogramming with small molecules instead of exogenous transcription factors. Stem Cells Int. 2015;2015:794632. doi: 10.1155/2015/794632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Polo J.M., Anderssen E., Walsh R.M., Schwarz B.A., Nefzger C.M., Lim S.M., Borkent M., Apostolou E., Alaei S., Cloutier J. A molecular roadmap of reprogramming somatic cells into iPS cells. Cell. 2012;151:1617–1632. doi: 10.1016/j.cell.2012.11.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Garber K. RIKEN suspends first clinical trial involving induced pluripotent stem cells. Nat. Biotech. 2015;33:890–891. doi: 10.1038/nbt0915-890. [DOI] [PubMed] [Google Scholar]
  • 13.Lund R.J., Narva E., Lahesmaa R. Genetic and epigenetic stability of human pluripotent stem cells. Nat. Rev. Genet. 2012;13:732–744. doi: 10.1038/nrg3271. [DOI] [PubMed] [Google Scholar]
  • 14.Morris S.A. Direct lineage reprogramming via pioneer factors; a detour through developmental gene regulatory networks. Development. 2016;143:2696–2705. doi: 10.1242/dev.138263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Fort A., Yamada D., Hashimoto K., Koseki H., Carninci P. Nuclear transcriptome profiling of induced pluripotent stem cells and embryonic stem cells identify non-coding loci resistant to reprogramming. Cell Cycle. 2015;14:1148–1155. doi: 10.4161/15384101.2014.988031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Xu J., Du Y., Deng H. Direct lineage reprogramming: strategies, mechanisms, and applications. Cell Stem Cell. 2015;16:119–134. doi: 10.1016/j.stem.2015.01.013. [DOI] [PubMed] [Google Scholar]
  • 17.Judson R.L., Greve T.S., Parchem R.J., Blelloch R. MicroRNA-based discovery of barriers to dedifferentiation of fibroblasts to pluripotent stem cells. Nat. Struct. Mol. Biol. 2013;20:1227–1235. doi: 10.1038/nsmb.2665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pfaff N., Fiedler J., Holzmann A., Schambach A., Moritz T., Cantz T., Thum T. miRNA screening reveals a new miRNA family stimulating iPS cell generation via regulation of Meox2. EMBO Rep. 2011;12:1153–1159. doi: 10.1038/embor.2011.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lee R.C., Feinbaum R.L., Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843–854. doi: 10.1016/0092-8674(93)90529-y. [DOI] [PubMed] [Google Scholar]
  • 20.Wightman B., Ha I., Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75:855–862. doi: 10.1016/0092-8674(93)90530-4. [DOI] [PubMed] [Google Scholar]
  • 21.Bartel D.P. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Winter J., Jung S., Keller S., Gregory R.I., Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat. Cell Biol. 2009;11:228–234. doi: 10.1038/ncb0309-228. [DOI] [PubMed] [Google Scholar]
  • 23.Gregory R.I., Yan K.P., Amuthan G., Chendrimada T., Doratotaj B., Cooch N., Shiekhattar R. The Microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432:235–240. doi: 10.1038/nature03120. [DOI] [PubMed] [Google Scholar]
  • 24.Han J., Lee Y., Yeom K.H., Kim Y.K., Jin H., Kim V.N. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004;18:3016–3027. doi: 10.1101/gad.1262504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lee Y., Ahn C., Han J., Choi H., Kim J., Yim J., Lee J., Provost P., Radmark O., Kim S. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003;425:415–419. doi: 10.1038/nature01957. [DOI] [PubMed] [Google Scholar]
  • 26.Ul Hussain M. Micro-RNAs (miRNAs): genomic organisation, biogenesis and mode of action. Cell Tissue Res. 2012;349:405–413. doi: 10.1007/s00441-012-1438-0. [DOI] [PubMed] [Google Scholar]
  • 27.Cheloufi S., Dos Santos C.O., Chong M.M., Hannon G.J. A dicer-independent miRNA biogenesis pathway that requires Ago catalysis. Nature. 2010;465:584–589. doi: 10.1038/nature09092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cifuentes D., Xue H., Taylor D.W., Patnode H., Mishima Y., Cheloufi S., Ma E., Mane S., Hannon G.J., Lawson N.D. A novel miRNA processing pathway independent of Dicer requires Argonaute2 catalytic activity. Science. 2010;328:1694–1698. doi: 10.1126/science.1190809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yates L.A., Norbury C.J., Gilbert R.J. The long and short of microRNA. Cell. 2013;153:516–519. doi: 10.1016/j.cell.2013.04.003. [DOI] [PubMed] [Google Scholar]
  • 30.Shan S.W., Fang L., Shatseva T., Rutnam Z.J., Yang X., Du W., Lu W.Y., Xuan J.W., Deng Z., Yang B.B. Mature miR-17-5p and passenger miR-17-3p induce hepatocellular carcinoma by targeting PTEN, GalNT7 and vimentin in different signal pathways. J. Cell Sci. 2013;126:1517–1530. doi: 10.1242/jcs.122895. [DOI] [PubMed] [Google Scholar]
  • 31.Yang X., Du W.W., Li H., Liu F., Khorshidi A., Rutnam Z.J., Yang B.B. Both mature miR-17-5p and passenger strand miR-17-3p target TIMP3 and induce prostate tumor growth and invasion. Nucleic Acids Res. 2013;41:9688–9704. doi: 10.1093/nar/gkt680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Giraldez A.J., Mishima Y., Rihel J., Grocock R.J., Van Dongen S., Inoue K., Enright A.J., Schier A.F. Zebrafish MiR-430 promotes deadenylation and clearance of maternal mRNAs. Science. 2006;312:75–79. doi: 10.1126/science.1122689. [DOI] [PubMed] [Google Scholar]
  • 33.HafezQorani S., Lafzi A., de Bruin R.G., van Zonneveld A.J., van der Veer E.P., Son Y.A., Kazan H. Modeling the combined effect of RNA-binding proteins and microRNAs in post-transcriptional regulation. Nucleic Acids Res. 2016;44:e83. doi: 10.1093/nar/gkw048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Humphreys D.T., Westman B.J., Martin D.I., Preiss T. MicroRNAs control translation initiation by inhibiting eukaryotic initiation factor 4E/cap and poly(A) tail function. Proc. Natl. Acad. Sci. U. S. A. 2005;102:16961–16966. doi: 10.1073/pnas.0506482102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Maroney P.A., Yu Y., Fisher J., Nilsen T.W. Evidence that microRNAs are associated with translating messenger RNAs in human cells. Nat. Struct. Mol. Biol. 2006;13:1102–1107. doi: 10.1038/nsmb1174. [DOI] [PubMed] [Google Scholar]
  • 36.Mathonnet G., Fabian M.R., Svitkin Y.V., Parsyan A., Huck L., Murata T., Biffo S., Merrick W.C., Darzynkiewicz E., Pillai R.S. MicroRNA inhibition of translation initiation in vitro by targeting the cap-binding complex eIF4F. Science. 2007;317:1764–1767. doi: 10.1126/science.1146067. [DOI] [PubMed] [Google Scholar]
  • 37.Nottrott S., Simard M.J., Richter J.D. Human let-7a miRNA blocks protein production on actively translating polyribosomes. Nat. Struct. Mol. Biol. 2006;13:1108–1114. doi: 10.1038/nsmb1173. [DOI] [PubMed] [Google Scholar]
  • 38.Pillai R.S., Bhattacharyya S.N., Artus C.G., Zoller T., Cougot N., Basyuk E., Bertrand E., Filipowicz W. Inhibition of translational initiation by Let-7 MicroRNA in human cells. Science. 2005;309:1573–1576. doi: 10.1126/science.1115079. [DOI] [PubMed] [Google Scholar]
  • 39.Wu L., Fan J., Belasco J.G. MicroRNAs direct rapid deadenylation of mRNA. Proc. Natl. Acad. Sci. U. S. A. 2006;103:4034–4039. doi: 10.1073/pnas.0510928103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chickarmane V., Troein C., Nuber U.A., Sauro H.M., Peterson C. Transcriptional dynamics of the embryonic stem cell switch. PLoS Comput. Biol. 2006;2:e123. doi: 10.1371/journal.pcbi.0020123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ebert M.S., Sharp P.A. Roles for microRNAs in conferring robustness to biological processes. Cell. 2012;149:515–524. doi: 10.1016/j.cell.2012.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Marson A., Levine S.S., Cole M.F., Frampton G.M., Brambrink T., Johnstone S., Guenther M.G., Johnston W.K., Wernig M., Newman J. Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell. 2008;134:521–533. doi: 10.1016/j.cell.2008.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Pujato M., MacCarthy T., Fiser A., Bergman A. The underlying molecular and network level mechanisms in the evolution of robustness in gene regulatory networks. PLoS Comput. Biol. 2013;9:e1002865. doi: 10.1371/journal.pcbi.1002865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gonzalez F., Boue S., Izpisua Belmonte J.C. Methods for making induced pluripotent stem cells: reprogramming a la carte. Nat. Rev. Genet. 2011;12:231–242. doi: 10.1038/nrg2937. [DOI] [PubMed] [Google Scholar]
  • 45.Li M., Belmonte J.C.I. Ground rules of the pluripotency gene regulatory network. Nat. Rev. Genet. 2017;18(3):180–191. doi: 10.1038/nrg.2016.156. advance online publication. [DOI] [PubMed] [Google Scholar]
  • 46.Ng H.H., Surani M.A. The transcriptional and signalling networks of pluripotency. Nat. Cell Biol. 2011;13:490–496. doi: 10.1038/ncb0511-490. [DOI] [PubMed] [Google Scholar]
  • 47.Rackham O.J., Firas J., Fang H., Oates M.E., Holmes M.L., Knaupp A.S., Consortium F., Suzuki H., Nefzger C.M., Daub C.O. A predictive computational framework for direct reprogramming between human cell types. Nat. Genet. 2016;48:331–335. doi: 10.1038/ng.3487. [DOI] [PubMed] [Google Scholar]
  • 48.Chen J., Wang G., Lu C., Guo X., Hong W., Kang J., Wang J. Synergetic cooperation of microRNAs with transcription factors in iPS cell generation. PLoS One. 2012;7:e40849. doi: 10.1371/journal.pone.0040849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Judson R.L., Babiarz J.E., Venere M., Blelloch R. Embryonic stem cell-specific microRNAs promote induced pluripotency. Nat. Biotechnol. 2009;27:459–461. doi: 10.1038/nbt.1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li Z., Dang J., Chang K.Y., Rana T.M. MicroRNA-mediated regulation of extracellular matrix formation modulates somatic cell reprogramming. RNA. 2014;20:1900–1915. doi: 10.1261/rna.043745.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Li Z., Yang C.S., Nakashima K., Rana T.M. Small RNA-mediated regulation of iPS cell generation. EMBO J. 2011;30:823–834. doi: 10.1038/emboj.2011.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lu D., Davis M.P., Abreu-Goodger C., Wang W., Campos L.S., Siede J., Vigorito E., Skarnes W.C., Dunham I., Enright A.J. MiR-25 regulates Wwp2 and Fbxw7 and promotes reprogramming of mouse fibroblast cells to iPSCs. PLoS One. 2012;7:e40938. doi: 10.1371/journal.pone.0040938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Miyoshi N., Ishii H., Nagano H., Haraguchi N., Dewi D.L., Kano Y., Nishikawa S., Tanemura M., Mimori K., Tanaka F. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8:633–638. doi: 10.1016/j.stem.2011.05.001. [DOI] [PubMed] [Google Scholar]
  • 54.Polo J.M., Anderssen E., Walsh R.M., Schwarz B.A., Nefzger C.M., Lim S.M., Borkent M., Apostolou E., Alaei S., Cloutier J. A molecular roadmap of reprogramming somatic cells into iPS cells. Cell. 2012;151:1617–1632. doi: 10.1016/j.cell.2012.11.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ruiz S., Panopoulos A.D., Montserrat N., Multon M.C., Daury A., Rocher C., Spanakis E., Batchelder E.M., Orsini C., Deleuze J.F. Generation of a drug-inducible reporter system to study cell reprogramming in human cells. J. Biol. Chem. 2012;287:40767–40778. doi: 10.1074/jbc.M112.384024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Samavarchi-Tehrani P., Golipour A., David L., Sung H.K., Beyer T.A., Datti A., Woltjen K., Nagy A., Wrana J.L. Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell. 2010;7:64–77. doi: 10.1016/j.stem.2010.04.015. [DOI] [PubMed] [Google Scholar]
  • 57.Wang G., Guo X., Hong W., Liu Q., Wei T., Lu C., Gao L., Ye D., Zhou Y., Chen J. Critical regulation of miR-200/ZEB2 pathway in Oct4/Sox2-induced mesenchymal-to-epithelial transition and induced pluripotent stem cell generation. Proc. Natl. Acad. Sci. U. S. A. 2013;110:2858–2863. doi: 10.1073/pnas.1212769110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wang J., He Q., Han C., Gu H., Jin L., Li Q., Mei Y., Wu M. p53-facilitated miR-199a-3p regulates somatic cell reprogramming. Stem Cells. 2012;30:1405–1413. doi: 10.1002/stem.1121. [DOI] [PubMed] [Google Scholar]
  • 59.Yang C.S., Li Z., Rana T.M. microRNAs modulate iPS cell generation. RNA. 2011;17:1451–1460. doi: 10.1261/rna.2664111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kim J., Chu J., Shen X., Wang J., Orkin S.H. An extended transcriptional network for pluripotency of embryonic stem cells. Cell. 2008;132:1049–1061. doi: 10.1016/j.cell.2008.02.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Subramanyam D., Lamouille S., Judson R.L., Liu J.Y., Bucay N., Derynck R., Blelloch R. Multiple targets of miR-302 and miR-372 promote reprogramming of human fibroblasts to induced pluripotent stem cells. Nat. Biotechnol. 2011;29:443–448. doi: 10.1038/nbt.1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Banito A., Rashid S.T., Acosta J.C., Li S., Pereira C.F., Geti I., Pinho S., Silva J.C., Azuara V., Walsh M. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 2009;23:2134–2139. doi: 10.1101/gad.1811609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.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. doi: 10.1038/nature08235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kawamura T., Suzuki J., Wang Y.V., Menendez S., Morera L.B., Raya A., Wahl G.M., Izpisua Belmonte J.C. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature. 2009;460:1140–1144. doi: 10.1038/nature08311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Marion R.M., Strati K., Li H., Murga M., Blanco R., Ortega S., Fernandez-Capetillo O., Serrano M., Blasco M.A. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature. 2009;460:1149–1153. doi: 10.1038/nature08287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lin C.P., Choi Y.J., Hicks G.G., He L. The emerging functions of the p53-miRNA network in stem cell biology. Cell Cycle. 2012;11:2063–2072. doi: 10.4161/cc.20207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Choi Y.J., Lin C.P., Ho J.J., He X., Okada N., Bu P., Zhong Y., Kim S.Y., Bennett M.J., Chen C. miR-34 miRNAs provide a barrier for somatic cell reprogramming. Nat. Cell Biol. 2011;13:1353–1360. doi: 10.1038/ncb2366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Jain A.K., Allton K., Iacovino M., Mahen E., Milczarek R.J., Zwaka T.P., Kyba M., Barton M.C. p53 regulates cell cycle and microRNAs to promote differentiation of human embryonic stem cells. PLoS Biol. 2012;10:e1001268. doi: 10.1371/journal.pbio.1001268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Xu N., Papagiannakopoulos T., Pan G., Thomson J.A., Kosik K.S. MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell. 2009;137:647–658. doi: 10.1016/j.cell.2009.02.038. [DOI] [PubMed] [Google Scholar]
  • 70.Gingold J.A., Fidalgo M., Guallar D., Lau Z., Sun Z., Zhou H., Faiola F., Huang X., Lee D.F., Waghray A. A genome-wide RNAi screen identifies opposing functions of Snai1 and Snai2 on the Nanog dependency in reprogramming. Mol. Cell. 2014;56:140–152. doi: 10.1016/j.molcel.2014.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ye D., Wang G., Liu Y., Huang W., Wu M., Zhu S., Jia W., Deng A.M., Liu H., Kang J. MiR-138 promotes induced pluripotent stem cell generation through the regulation of the p53 signaling. Stem Cells. 2012;30:1645–1654. doi: 10.1002/stem.1149. [DOI] [PubMed] [Google Scholar]
  • 72.Hysolli E., Tanaka Y., Su J., Kim K.Y., Zhong T., Janknecht R., Zhou X.L., Geng L., Qiu C., Pan X. Regulation of the DNA methylation landscape in human somatic cell reprogramming by the miR-29 family. Stem Cell Rep. 2016;7:43–54. doi: 10.1016/j.stemcr.2016.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Guo X., Liu Q., Wang G., Zhu S., Gao L., Hong W., Chen Y., Wu M., Liu H., Jiang C. microRNA-29b is a novel mediator of Sox2 function in the regulation of somatic cell reprogramming. Cell Res. 2013;23:142–156. doi: 10.1038/cr.2012.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Niu M., Gao D., Wen Q., Wei P., Pan S., Shuai C., Ma H., Xiang J., Li Z., Fan S. MiR-29c regulates the expression of miR-34c and miR-449a by targeting DNA methyltransferase 3a and 3b in nasopharyngeal carcinoma. BMC Cancer. 2016;16:218. doi: 10.1186/s12885-016-2253-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pandi G., Nakka V.P., Dharap A., Roopra A., Vemuganti R. MicroRNA miR-29c down-regulation leading to de-repression of its target DNA methyltransferase 3a promotes ischemic brain damage. PLoS One. 2013;8:e58039. doi: 10.1371/journal.pone.0058039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Yang G., Song Y., Zhou X., Deng Y., Liu T., Weng G., Yu D., Pan S. DNA methyltransferase 3, a target of microRNA-29c, contributes to neuronal proliferation by regulating the expression of brain-derived neurotrophic factor. Mol. Med. Rep. 2015;12:1435–1442. doi: 10.3892/mmr.2015.3531. [DOI] [PubMed] [Google Scholar]
  • 77.Card D.A., Hebbar P.B., Li L., Trotter K.W., Komatsu Y., Mishina Y., Archer T.K. Oct4/Sox2-regulated miR-302 targets cyclin D1 in human embryonic stem cells. Mol. Cell Biol. 2008;28:6426–6438. doi: 10.1128/MCB.00359-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Anokye-Danso F., Trivedi C.M., Juhr D., Gupta M., Cui Z., Tian Y., Zhang Y., Yang W., Gruber P.J., Epstein J.A. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 2011;8:376–388. doi: 10.1016/j.stem.2011.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Konno M., Koseki J., Kawamoto K., Nishida N., Matsui H., Dewi D.L., Ozaki M., Noguchi Y., Mimori K., Gotoh N. Embryonic microRNA-369 controls metabolic splicing factors and urges cellular reprograming. PLoS One. 2015;10:e0132789. doi: 10.1371/journal.pone.0132789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Tanabe K., Haag D., Wernig M. Direct somatic lineage conversion. Philos. Trans. R. Soc. Lond B Biol. Sci. 2015;370:20140368. doi: 10.1098/rstb.2014.0368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Vierbuchen T., Wernig M. Direct lineage conversions: unnatural but useful? Nat. Biotechnol. 2011;29:892–907. doi: 10.1038/nbt.1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.An N., Xu H., Gao W.Q., Yang H. Direct conversion of somatic cells into induced neurons. Mol. Neurobiol. 2016 doi: 10.1007/s12035-016-0350-0. [DOI] [PubMed] [Google Scholar]
  • 83.Spater D., Hansson E.M., Zangi L., Chien K.R. How to make a cardiomyocyte. Development. 2014;141:4418–4431. doi: 10.1242/dev.091538. [DOI] [PubMed] [Google Scholar]
  • 84.Abdullah A.I., Pollock A., Sun T. The path from skin to brain: generation of functional neurons from fibroblasts. Mol. Neurobiol. 2012;45:586–595. doi: 10.1007/s12035-012-8277-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Amamoto R., Arlotta P. Reshaping the brain: direct lineage conversion in the nervous system. F1000Prime Rep. 2013;5:33. doi: 10.12703/P5-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Arlotta P., Berninger B. Brains in metamorphosis: reprogramming cell identity within the central nervous system. Curr. Opin. Neurobiol. 2014;27:208–214. doi: 10.1016/j.conb.2014.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Davis-Dusenbery B.N., Williams L.A., Klim J.R., Eggan K. How to make spinal motor neurons. Development. 2014;141:491–501. doi: 10.1242/dev.097410. [DOI] [PubMed] [Google Scholar]
  • 88.Doppler S.A., Deutsch M.A., Lange R., Krane M. Direct reprogramming-the future of cardiac regeneration? Int. J. Mol. Sci. 2015;16:17368–17393. doi: 10.3390/ijms160817368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Huang Y., Tan S. Direct lineage conversion of astrocytes to induced neural stem cells or neurons. Neurosci. Bull. 2015;31:357–367. doi: 10.1007/s12264-014-1517-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ieda M., Fukuda K. Cardiomyocyte generation using stem cells and directly reprogrammed cells. Front. Biosci. Sch. Ed. 2012;4:1413–1423. doi: 10.2741/s343. [DOI] [PubMed] [Google Scholar]
  • 91.Kim J., Ambasudhan R., Ding S. Direct lineage reprogramming to neural cells. Curr. Opin. Neurobiol. 2012;22:778–784. doi: 10.1016/j.conb.2012.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Masip M., Veiga A., Izpisua Belmonte J.C., Simon C. Reprogramming with defined factors: from induced pluripotency to induced transdifferentiation. Mol. Hum. Reprod. 2010;16:856–868. doi: 10.1093/molehr/gaq059. [DOI] [PubMed] [Google Scholar]
  • 93.Masserdotti G., Gascon S., Gotz M. Direct neuronal reprogramming: learning from and for development. Development. 2016;143:2494–2510. doi: 10.1242/dev.092163. [DOI] [PubMed] [Google Scholar]
  • 94.Morris S.A., Daley G.Q. A blueprint for engineering cell fate: current technologies to reprogram cell identity. Cell Res. 2013;23:33–48. doi: 10.1038/cr.2013.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Roessler R., Boddeke E., Copray S. Induced pluripotent stem cell technology and direct conversion: new possibilities to study and treat Parkinson's disease. Stem Cell Rev. 2013;9:505–513. doi: 10.1007/s12015-012-9369-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sancho-Martinez I., Baek S.H., Izpisua Belmonte J.C. Lineage conversion methodologies meet the reprogramming toolbox. Nat. Cell Biol. 2012;14:892–899. doi: 10.1038/ncb2567. [DOI] [PubMed] [Google Scholar]
  • 97.Shi Z., Jiao J. Direct lineage conversion: induced neuronal cells and induced neural stem cells. Protein Cell. 2012;3:826–833. doi: 10.1007/s13238-012-2068-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Velasco I., Salazar P., Giorgetti A., Ramos-Mejia V., Castano J., Romero-Moya D., Menendez P. Concise review: generation of neurons from somatic cells of healthy individuals and neurological patients through induced pluripotency or direct conversion. Stem Cells. 2014;32:2811–2817. doi: 10.1002/stem.1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Yoo A.S., Staahl B.T., Chen L., Crabtree G.R. MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature. 2009;460:642–646. doi: 10.1038/nature08139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Yoo A.S., Sun A.X., Li L., Shcheglovitov A., Portmann T., Li Y., Lee-Messer C., Dolmetsch R.E., Tsien R.W., Crabtree G.R. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature. 2011;476:228–231. doi: 10.1038/nature10323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Victor M.B., Richner M., Hermanstyne T.O., Ransdell J.L., Sobieski C., Deng P.Y., Klyachko V.A., Nerbonne J.M., Yoo A.S. Generation of human striatal neurons by microRNA-dependent direct conversion of fibroblasts. Neuron. 2014;84:311–323. doi: 10.1016/j.neuron.2014.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ballas N., Grunseich C., Lu D.D., Speh J.C., Mandel G. REST and its corepressors mediate plasticity of neuronal gene chromatin throughout neurogenesis. Cell. 2005;121:645–657. doi: 10.1016/j.cell.2005.03.013. [DOI] [PubMed] [Google Scholar]
  • 103.Conaco C., Otto S., Han J.J., Mandel G. Reciprocal actions of REST and a microRNA promote neuronal identity. Proc. Natl. Acad. Sci. U. S. A. 2006;103:2422–2427. doi: 10.1073/pnas.0511041103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Xue Y., Ouyang K., Huang J., Zhou Y., Ouyang H., Li H., Wang G., Wu Q., Wei C., Bi Y. Direct conversion of fibroblasts to neurons by reprogramming PTB-regulated microRNA circuits. Cell. 2013;152:82–96. doi: 10.1016/j.cell.2012.11.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Packer A.N., Xing Y., Harper S.Q., Jones L., Davidson B.L. The bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and is downregulated in Huntington's disease. J. Neurosci. 2008;28:14341–14346. doi: 10.1523/JNEUROSCI.2390-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Selbach M., Schwanhausser B., Thierfelder N., Fang Z., Khanin R., Rajewsky N. Widespread changes in protein synthesis induced by microRNAs. Nature. 2008;455:58–63. doi: 10.1038/nature07228. [DOI] [PubMed] [Google Scholar]
  • 107.Pang Z.P., Yang N., Vierbuchen T., Ostermeier A., Fuentes D.R., Yang T.Q., Citri A., Sebastiano V., Marro S., Sudhof T.C. Induction of human neuronal cells by defined transcription factors. Nature. 2011;476:220–223. doi: 10.1038/nature10202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jayawardena T.M., Egemnazarov B., Finch E.A., Zhang L., Payne J.A., Pandya K., Zhang Z., Rosenberg P., Mirotsou M., Dzau V.J. MicroRNA-mediated in vitro and in vivo direct reprogramming of cardiac fibroblasts to cardiomyocytes. Circ. Res. 2012;110:1465–1473. doi: 10.1161/CIRCRESAHA.112.269035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wang X., Hodgkinson C.P., Lu K., Payne A.J., Pratt R.E., Dzau V.J. Selenium augments microRNA directed reprogramming of fibroblasts to cardiomyocytes via Nanog. Sci. Rep. 2016;6:23017. doi: 10.1038/srep23017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Muraoka N., Yamakawa H., Miyamoto K., Sadahiro T., Umei T., Isomi M., Nakashima H., Akiyama M., Wada R., Inagawa K. MiR-133 promotes cardiac reprogramming by directly repressing Snai1 and silencing fibroblast signatures. EMBO J. 2014;33:1565–1581. doi: 10.15252/embj.201387605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ghasemi-Kasman M., Hajikaram M., Baharvand H., Javan M. MicroRNA-mediated in vitro and in vivo direct conversion of astrocytes to neuroblasts. PLoS One. 2015;10:e0127878. doi: 10.1371/journal.pone.0127878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Zhou C., Gu H., Fan R., Wang B., Lou J. MicroRNA 302/367 cluster effectively facilitates direct reprogramming from human fibroblasts into functional neurons. Stem Cells Dev. 2015;24:2746–2755. doi: 10.1089/scd.2015.0123. [DOI] [PubMed] [Google Scholar]
  • 113.Lu J., Dong H., Lin L., Wang Q., Huang L., Tan J. miRNA-302 facilitates reprogramming of human adult hepatocytes into pancreatic islets-like cells in combination with a chemical defined media. Biochem. Biophys. Res. Commun. 2014;453:405–410. doi: 10.1016/j.bbrc.2014.09.095. [DOI] [PubMed] [Google Scholar]
  • 114.Forrest A.R., Kawaji H., Rehli M., Baillie J.K., de Hoon M.J., Haberle V., Lassmann T., Kulakovskiy I.V., Lizio M., Itoh M. A promoter-level mammalian expression atlas. Nature. 2014;507:462–470. doi: 10.1038/nature13182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Consortium E.P. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.de Hoon M., Shin J.W., Carninci P. Paradigm shifts in genomics through the FANTOM projects. Mamm. Genome. 2015;26:391–402. doi: 10.1007/s00335-015-9593-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Quinn J.J., Chang H.Y. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 2016;17:47–62. doi: 10.1038/nrg.2015.10. [DOI] [PubMed] [Google Scholar]
  • 118.Signal B., Gloss B.S., Dinger M.E. Computational approaches for functional prediction and characterisation of long noncoding RNAs. Trends Genet. 2016;32:620–637. doi: 10.1016/j.tig.2016.08.004. [DOI] [PubMed] [Google Scholar]
  • 119.Wang K.C., Chang H.Y. Molecular mechanisms of long noncoding RNAs. Mol. Cell. 2011;43:904–914. doi: 10.1016/j.molcel.2011.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Chen L.L. Linking long noncoding RNA localization and function. Trends Biochem. Sci. 2016;41:761–772. doi: 10.1016/j.tibs.2016.07.003. [DOI] [PubMed] [Google Scholar]
  • 121.Johnsson P., Lipovich L., Grandér D., Morris K.V. Evolutionary conservation of long non-coding RNAs; sequence, structure, function. Biochimica Biophysica Acta (BBA) - General Subj. 2014;1840:1063–1071. doi: 10.1016/j.bbagen.2013.10.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chen L., Zhang S. Long noncoding RNAs in cell differentiation and pluripotency. Cell Tissue Res. 2016;366:509–521. doi: 10.1007/s00441-016-2451-5. [DOI] [PubMed] [Google Scholar]
  • 123.Dey B.K., Mueller A.C., Dutta A. Long non-coding RNAs as emerging regulators of differentiation, development, and disease. Transcription. 2014;5:e944014. doi: 10.4161/21541272.2014.944014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Fatica A., Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat. Rev. Genet. 2014;15:7–21. doi: 10.1038/nrg3606. [DOI] [PubMed] [Google Scholar]
  • 125.Flynn R.A., Chang H.Y. Long noncoding RNAs in cell-fate programming and reprogramming. Cell Stem Cell. 2014;14:752–761. doi: 10.1016/j.stem.2014.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ghosal S., Das S., Chakrabarti J. Long noncoding RNAs: new players in the molecular mechanism for maintenance and differentiation of pluripotent stem cells. Stem Cells Dev. 2013;22:2240–2253. doi: 10.1089/scd.2013.0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Guan D., Zhang W., Zhang W., Liu G.H., Belmonte J.C. Switching cell fate, ncRNAs coming to play. Cell Death Dis. 2013;4:e464. doi: 10.1038/cddis.2012.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hu S., Shan G. LncRNAs in stem cells. Stem Cells Int. 2016;2016:2681925. doi: 10.1155/2016/2681925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Hu W., Alvarez-Dominguez J.R., Lodish H.F. Regulation of mammalian cell differentiation by long non-coding RNAs. EMBO Rep. 2012;13:971–983. doi: 10.1038/embor.2012.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lopez-Pajares V. Long non-coding RNA regulation of gene expression during differentiation. Pflugers Arch. 2016;468:971–981. doi: 10.1007/s00424-016-1809-6. [DOI] [PubMed] [Google Scholar]
  • 131.Rosa A., Ballarino M. Long noncoding RNA regulation of pluripotency. Stem Cells Int. 2016;2016:1797692. doi: 10.1155/2016/1797692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Hussein S.M., Puri M.C., Tonge P.D., Benevento M., Corso A.J., Clancy J.L., Mosbergen R., Li M., Lee D.S., Cloonan N. Genome-wide characterization of the routes to pluripotency. Nature. 2014;516:198–206. doi: 10.1038/nature14046. [DOI] [PubMed] [Google Scholar]
  • 133.Loewer S., Cabili M.N., Guttman M., Loh Y.H., Thomas K., Park I.H., Garber M., Curran M., Onder T., Agarwal S. Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat. Genet. 2010;42:1113–1117. doi: 10.1038/ng.710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Wang Y., Xu Z., Jiang J., Xu C., Kang J., Xiao L., Wu M., Xiong J., Guo X., Liu H. Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev. Cell. 2013;25:69–80. doi: 10.1016/j.devcel.2013.03.002. [DOI] [PubMed] [Google Scholar]
  • 135.Bao X., Wu H., Zhu X., Guo X., Hutchins A.P., Luo Z., Song H., Chen Y., Lai K., Yin M. The p53-induced lincRNA-p21 derails somatic cell reprogramming by sustaining H3K9me3 and CpG methylation at pluripotency gene promoters. Cell Res. 2015;25:80–92. doi: 10.1038/cr.2014.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Jain A.K., Xi Y., McCarthy R., Allton K., Akdemir K.C., Patel L.R., Aronow B., Lin C., Li W., Yang L. LncPRESS1 is a p53-regulated LncRNA that safeguards pluripotency by disrupting SIRT6-mediated de-acetylation of histone H3K56. Mol. Cell. 2016;64:967–981. doi: 10.1016/j.molcel.2016.10.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Dinger M.E., Amaral P.P., Mercer T.R., Pang K.C., Bruce S.J., Gardiner B.B., Askarian-Amiri M.E., Ru K., Solda G., Simons C. Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res. 2008;18:1433–1445. doi: 10.1101/gr.078378.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ng S.Y., Johnson R., Stanton L.W. Human long non-coding RNAs promote pluripotency and neuronal differentiation by association with chromatin modifiers and transcription factors. EMBO J. 2012;31:522–533. doi: 10.1038/emboj.2011.459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Sheik Mohamed J., Gaughwin P.M., Lim B., Robson P., Lipovich L. Conserved long noncoding RNAs transcriptionally regulated by Oct4 and Nanog modulate pluripotency in mouse embryonic stem cells. RNA. 2010;16:324–337. doi: 10.1261/rna.1441510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Guttman M., Donaghey J., Carey B.W., Garber M., Grenier J.K., Munson G., Young G., Lucas A.B., Ach R., Bruhn L. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature. 2011;477:295–300. doi: 10.1038/nature10398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Chakraborty D., Kappei D., Theis M., Nitzsche A., Ding L., Paszkowski-Rogacz M., Surendranath V., Berger N., Schulz H., Saar K. Combined RNAi and localization for functionally dissecting long noncoding RNAs. Nat. Meth. 2012;9:360–362. doi: 10.1038/nmeth.1894. [DOI] [PubMed] [Google Scholar]
  • 142.Liu S.J., Horlbeck M.A., Cho S.W., Birk H.S., Malatesta M., He D., Attenello F.J., Villalta J.E., Cho M.Y., Chen Y. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science. 2017;355(6320) doi: 10.1126/science.aah7111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Margueron R., Reinberg D. The Polycomb complex PRC2 and its mark in life. Nature. 2011;469:343–349. doi: 10.1038/nature09784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kaneko S., Bonasio R., Saldana-Meyer R., Yoshida T., Son J., Nishino K., Umezawa A., Reinberg D. Interactions between JARID2 and noncoding RNAs regulate PRC2 recruitment to chromatin. Mol. Cell. 2014;53:290–300. doi: 10.1016/j.molcel.2013.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Savic N., Bar D., Leone S., Frommel S.C., Weber F.A., Vollenweider E., Ferrari E., Ziegler U., Kaech A., Shakhova O. lncRNA maturation to initiate heterochromatin formation in the nucleolus is required for exit from pluripotency in ESCs. Cell Stem Cell. 2014;15:720–734. doi: 10.1016/j.stem.2014.10.005. [DOI] [PubMed] [Google Scholar]
  • 146.Lu X., Sachs F., Ramsay L., Jacques P.E., Goke J., Bourque G., Ng H.H. The retrovirus HERVH is a long noncoding RNA required for human embryonic stem cell identity. Nat. Struct. Mol. Biol. 2014;21:423–425. doi: 10.1038/nsmb.2799. [DOI] [PubMed] [Google Scholar]
  • 147.Lin N., Chang K.Y., Li Z., Gates K., Rana Z.A., Dang J., Zhang D., Han T., Yang C.S., Cunningham T.J. An evolutionarily conserved long noncoding RNA TUNA controls pluripotency and neural lineage commitment. Mol. Cell. 2014;53:1005–1019. doi: 10.1016/j.molcel.2014.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Zhou Y., Dai Q.S., Zhu S.C., Han Y.H., Han H.L., Zhao B., Gao R.R., Zhang J., Zhang J. AK048794 maintains the mouse embryonic stem cell pluripotency by functioning as an miRNA sponge for miR-592. Biochem. J. 2016;473:3639–3654. doi: 10.1042/BCJ20160540. [DOI] [PubMed] [Google Scholar]
  • 149.Xu C., Zhang Y., Wang Q., Xu Z., Jiang J., Gao Y., Gao M., Kang J., Wu M., Xiong J. Long non-coding RNA GAS5 controls human embryonic stem cell self-renewal by maintaining NODAL signalling. Nat. Commun. 2016;7:13287. doi: 10.1038/ncomms13287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Roberts T.C., Morris K.V., Wood M.J. The role of long non-coding RNAs in neurodevelopment, brain function and neurological disease. Philos. Trans. R. Soc. Lond B Biol. Sci. 2014;369 doi: 10.1098/rstb.2013.0507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ng S.Y., Bogu G.K., Soh B.S., Stanton L.W. The long noncoding RNA RMST interacts with SOX2 to regulate neurogenesis. Mol. Cell. 2013;51:349–359. doi: 10.1016/j.molcel.2013.07.017. [DOI] [PubMed] [Google Scholar]
  • 152.Lin M., Pedrosa E., Shah A., Hrabovsky A., Maqbool S., Zheng D., Lachman H.M. RNA-Seq of human neurons derived from iPS cells reveals candidate long non-coding RNAs involved in neurogenesis and neuropsychiatric disorders. PLoS One. 2011;6:e23356. doi: 10.1371/journal.pone.0023356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Mo C.-F., Wu F.-C., Tai K.-Y., Chang W.-C., Chang K.-W., Kuo H.-C., Ho H.-N., Chen H.-F., Lin S.-P. Loss of non-coding RNA expression from the DLK1-DIO3 imprinted locus correlates with reduced neural differentiation potential in human embryonic stem cell lines. Stem Cell Res. Ther. 2015;6:1. doi: 10.1186/scrt535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Qin H., Zhao A., Zhang C., Fu X. Epigenetic control of reprogramming and transdifferentiation by histone modifications. Stem Cell Rev. 2016;12:708–720. doi: 10.1007/s12015-016-9682-4. [DOI] [PubMed] [Google Scholar]
  • 155.Faridani O.R., Abdullayev I., Hagemann-Jensen M., Schell J.P., Lanner F., Sandberg R. Single-cell sequencing of the small-RNA transcriptome. Nat. Biotechnol. 2016;34:1264–1266. doi: 10.1038/nbt.3701. [DOI] [PubMed] [Google Scholar]
  • 156.Kim D.H., Marinov G.K., Pepke S., Singer Z.S., He P., Williams B., Schroth G.P., Elowitz M.B., Wold B.J. Single-cell transcriptome analysis reveals dynamic changes in lncRNA expression during reprogramming. Cell Stem Cell. 2015;16:88–101. doi: 10.1016/j.stem.2014.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Non-coding RNA Research are provided here courtesy of KeAi Publishing

RESOURCES