Embryonic Stem Cell Self-Renewal Pathways Converge on the Transcription Factor Tfcp2l1
Shoudong Ye,Ping Li,Chang Tong,Qi‐Long Ying
DOI: https://doi.org/10.1038/emboj.2013.175
2013-01-01
Abstract:Article13 August 2013free access Source Data Embryonic stem cell self-renewal pathways converge on the transcription factor Tfcp2l1 Shoudong Ye Shoudong Ye Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Department of Biochemistry and Molecular Biology, Shanghai Medical School and Key Laboratory of Molecular Medicine, Ministry of Education, Fudan University, Shanghai, PR China Search for more papers by this author Ping Li Ping Li Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Search for more papers by this author Chang Tong Chang Tong Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Search for more papers by this author Qi-Long Ying Corresponding Author Qi-Long Ying Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Search for more papers by this author Shoudong Ye Shoudong Ye Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Department of Biochemistry and Molecular Biology, Shanghai Medical School and Key Laboratory of Molecular Medicine, Ministry of Education, Fudan University, Shanghai, PR China Search for more papers by this author Ping Li Ping Li Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Search for more papers by this author Chang Tong Chang Tong Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Search for more papers by this author Qi-Long Ying Corresponding Author Qi-Long Ying Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA Search for more papers by this author Author Information Shoudong Ye1,2, Ping Li1, Chang Tong1 and Qi-Long Ying 1 1Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA 2Department of Biochemistry and Molecular Biology, Shanghai Medical School and Key Laboratory of Molecular Medicine, Ministry of Education, Fudan University, Shanghai, PR China *Corresponding author. Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, 1425 San Pablo Street, BCC 512, Los Angeles, CA 90033, USA. Tel.:+1 323 442 3308; Fax:+1 323 442 4040; E-mail: [email protected] The EMBO Journal (2013)32:2548-2560https://doi.org/10.1038/emboj.2013.175 There is a Have you seen? (October 2013) associated with this Article. PDFDownload PDF of article text and main figures. Peer ReviewDownload a summary of the editorial decision process including editorial decision letters, reviewer comments and author responses to feedback. ToolsAdd to favoritesDownload CitationsTrack CitationsPermissions ShareFacebookTwitterLinked InMendeleyWechatReddit Figures & Info Mouse embryonic stem cell (mESC) self-renewal can be maintained by activation of the leukaemia inhibitory factor (LIF)/signal transducer and activator of transcription 3 (Stat3) signalling pathway or dual inhibition (2i) of glycogen synthase kinase 3 (Gsk3) and mitogen-activated protein kinase kinase (MEK). Several downstream targets of the pathways involved have been identified that when individually overexpressed can partially support self-renewal. However, none of these targets is shared among the involved pathways. Here, we show that the CP2 family transcription factor Tfcp2l1 is a common target in LIF/Stat3- and 2i-mediated self-renewal, and forced expression of Tfcp2l1 can recapitulate the self-renewal-promoting effect of LIF or either of the 2i components. In addition, Tfcp2l1 can reprogram post-implantation epiblast stem cells to naïve pluripotent ESCs. Tfcp2l1 upregulates Nanog expression and promotes self-renewal in a Nanog-dependent manner. We conclude that Tfcp2l1 is at the intersection of LIF- and 2i-mediated self-renewal pathways and plays a critical role in maintaining ESC identity. Our study provides an expanded understanding of the current model of ground-state pluripotency. Introduction Embryonic stem cells (ESCs) are pluripotent cells derived from the inner cell mass of the pre-implantation blastocyst (Smith, 2001). ESCs were first established from mice (Evans and Kaufman, 1981; Martin, 1981), and more recently, from rats (Buehr et al, 2008; Li et al, 2008). Attempts to derive authentic ESCs from other mammals have so far failed, highlighting the need to understand more about how the ESC state is established and maintained. Mouse ESC (mESC) self-renewal can be maintained in two distinct culture systems: serum-containing medium supplemented with leukaemia inhibitor factor (LIF) (Smith et al, 1988; Williams et al, 1988), and serum-free N2B27 medium supplemented with two small molecule inhibitors (2i), CHIR99021 (CHIR) and PD0325901 (PD03) (Ying et al, 2008). LIF supports self-renewal by inducing activation of signal transducer and activator of transcription 3 (Stat3) (Niwa et al, 1998). CHIR and PD03 maintain self-renewal through inhibition of glycogen synthase kinase 3 (Gsk3) and mitogen-activated protein kinase kinase (MEK), respectively. When overexpressed, several LIF/Stat3 downstream targets, including Klf4, Gbx2, Pim1, Pim3, Pramel7, Rhox5, and c-Myc, can partially recapitulate the self-renewal-promoting effect of LIF/Stat3 signalling (Cartwright et al, 2005; Aksoy et al, 2007; Cinelli et al, 2008; Bourillot et al, 2009; Hall et al, 2009; Casanova et al, 2011; Tai and Ying, 2013). Knockdown of any of those Stat3 downstream targets, however, does not abolish LIF/Stat3-mediated mESC self-renewal, suggesting that LIF/Stat3 might activate multiple downstream targets with cross-compensatory functionality, ensuring the integrity of the self-renewal program. It is also possible that the master downstream targets of LIF/Stat3 have yet to be identified. CHIR inhibits Gsk3, leading to the stabilization and nuclear translocation of β-catenin and subsequent activation of canonical Wnt/β-catenin signalling. CHIR-induced activation of Wnt/β-catenin signalling abrogates T-cell factor 3 (Tcf3)-mediated repression of the pluripotency network, and this has been proposed to be a key mechanism underlying CHIR's role in promoting mESC self-renewal (Wray et al, 2011). One of the key targets of Tcf3 repression is Esrrb, whose expression is essential for mESC self-renewal mediated by inhibition of Gsk3 by CHIR (Martello et al, 2012). CHIR alone is not sufficient for maintaining mESC self-renewal, and addition of PD03 is required. PD03 enhances Nanog expression, which may account for the self-renewal-promoting effect of this small molecule (Miyanari and Torres-Padilla, 2012). Although LIF and 2i promote mESC self-renewal through seemingly independent pathways, they might converge on the same downstream targets to exert their self-renewal-promoting effect. However, no direct downstream connection has yet been established. LIF can replace PD03 or CHIR in the presence of either to recapitulate the combined self-renewal-sustaining effect of 2i in serum-free culture (Wray et al, 2010), indicating possible overlap in the self-renewal machinery activated by LIF and 2i. This notion is further supported by the facts that Stat3-null mESC self-renewal can be maintained by 2i (Ying et al, 2008), and LIF/PD03 is sufficient to maintain β-catenin-null ESCs that are not responsive to CHIR for self-renewal (Lyashenko et al, 2011). Uncovering how these molecular pathways overlap will provide further insight into not only how ESCs maintain their intrinsic properties, but also how stem cells may be exploited for medical and technological purposes. Such an advance might also facilitate the development of conditions for the derivation of authentic ESCs from species other than mice and rats. Here, we identified transcription factor CP2-like 1 (Tfcp2l1, also called Tcfcp2l1 or CRTR-1) as a common downstream target of LIF/Stat3, CHIR, and PD03, that when overexpressed, can recapitulate the self-renewal-promoting effect of each. Results Identification of genes differentially induced/suppressed in mESCs by LIF and LIF/2i Under feeder-free conditions, LIF alone is not sufficient to maintain self-renewal of ESCs derived from non-129 strains of mice such as C57BL/6 and BALB/c, and addition of 2i is required (Gertsenstein et al, 2010; Wray et al, 2010; Ye et al, 2012). We hypothesized that 2i promotes ESC self-renewal by inducing the expression of pluripotency genes insufficiently induced by LIF or through suppressing differentiation-inducing genes. We performed DNA microarray analysis to identify genes differentially expressed in C57BL/6 mESCs treated with LIF or LIF/2i. We identified 446 genes upregulated and 841 genes downregulated by 1.5-fold or greater in LIF/2i compared with LIF alone (GEO ID Number: GSE46369). We focussed on genes that are specifically expressed in pluripotent stem cells and are upregulated in the LIF/2i condition, and identified five candidates: Tbx3, Klf2, Klf4, Nanog, and Tfcp2l1 (Figure 1A). The differential expression levels of these candidate genes in LIF/2i and LIF alone were confirmed by quantitative real-time PCR (qRT–PCR) (Figure 1B). We then overexpressed these genes in C57BL/6 mESCs (Figure 1C), and all were able to support feeder-free C57BL/6 mESC self-renewal in the presence of LIF alone (Figure 1D and E). The results for Tbx3, Klf2, Klf4, and Nanog were consistent with previous reports that overexpression of these genes promoted self-renewal in 46C and E14 mESCs without exogenous LIF (Hall et al, 2009; Niwa et al, 2009). Tfcp2l1 is preferentially expressed in ESCs (Ivanova et al, 2006), but its function in ESC self-renewal has not been characterized. This prompted us to undertake further analysis of the role of Tfcp2l1 in mESC self-renewal. Figure 1.2i induces multiple transcription factors to promote mESC self-renewal. (A) Scatter plots of DNA microarray data showing Tfcp2l1, Klf2, Klf4, Nanog, and Tbx3 gene signal intensities for C57BL/6 mESCs cultured in serum/LIF/2i or serum/LIF for 12 h. (B) qRT–PCR analysis of Tbx3, Klf2, Klf4, Tfcp2l1, and Nanog expression in C57BL/6 mESCs cultured in the presence of serum/LIF/2i or serum/LIF for 12 h. Error bars are the s.d. of three biological replicates. (C) Western blot analysis of indicated Flag-tagged proteins in C57BL/6 mESCs cultured in LIF/2i. α-Tubulin is a loading control. (D) Alkaline phosphatase staining of colonies arising from transfectants cultured in serum/LIF for two passages. Scale bar, 100 μm. (E) Quantification of alkaline phosphatase-positive colonies. Data represent mean±s.d. from triplicate experiments.Source data for this figure is available on the online supplementary information page. Source Data for Figure 1C [embj2013175-sup-0001-SourceData-S1.pdf] Download figure Download PowerPoint Tfcp2l1 overexpression can substitute for PD03 or CHIR in 2i to maintain mESC self-renewal To determine whether Tfcp2l1 expression is induced by CHIR or PD03 or both, we analysed its expression in mESCs treated with one or both of these small molecules in the presence of LIF. Tfcp2l1 expression was induced in all three conditions (Figure 2A). To determine the function of Tfcp2l1 in mESC self-renewal, we used an inducible cassette exchange (ICE) system (Iacovino et al, 2011) to generate an mESC line harbouring a doxycycline (Dox)-inducible Tfcp2l1 transgene (i-Tfcp2l1 mESCs). Western blot analysis confirmed that Tfcp2l1 expression in these cells was efficiently induced by Dox treatment (Figure 2B). i-Tfcp2l1 mESCs as well as control mESCs harbouring a Dox-inducible EGFP transgene (i-EGFP mESCs) could be efficiently maintained in the serum-free N2B27/2i condition with or without Dox. Withdrawal of 2i resulted in cell death or differentiation within five passages even in the presence of Dox, suggesting that artificial Tfcp2l1 expression cannot replace 2i for the maintenance of mESC self-renewal. This prompted us to examine the relationship between Tfcp2l1 and each 2i component in maintaining mESC self-renewal. i-Tfcp2l1 mESCs could be continually passaged in N2B27/CHIR plus Dox while retaining typical ESC morphology and an alkaline phosphatase-positive staining profile (Figure 2C–E). qRT–PCR analysis revealed high-level expression of the pluripotency genes Klf4 and Nanog and low-level expression of genes associated with differentiation (Figure 2F), and immunofluorescence staining showed positive expression of the pluripotency markers Oct4 and SSEA1 (Supplementary Figure S1). Following the removal of Dox, i-Tfcp2l1 mESCs retained the capacity to differentiate into Nestin-positive neurons, Myosin-positive myocardial cells, and Gata4-positive primitive endoderm cells in vitro (Figure 2G). Notably, Dox-induced Tfcp2l1 expression did not alter the phosphorylation level of extracellular signal-regulated kinase (ERK), suggesting that the ability of Tfcp2l1 to replace PD03 is not attributable to inhibition of ERK phosphorylation (Supplementary Figure S2). In N2B27/PD03 plus Dox, undifferentiated i-Tfcp2l1 mESCs could also be continually passaged, although they proliferated more slowly than did i-Tfcp2l1 mESCs in CHIR or 2i (Figure 2H and I). Taken together, these findings demonstrate that Tfcp2l1 transcription can be upregulated by both CHIR and PD03, and that forced expression of Tfcp2l1 can recapitulate the effect of PD03, and partially that of CHIR, in promoting mESC self-renewal in the 2i condition. Figure 2.Enforced Tfcp2l1 phenocopies PD03 or CHIR to promote ground-state pluripotency. (A) qRT–PCR analysis of Tfcp2l1 expression in 46C mESCs treated for 12 h with LIF, CHIR, and/or PD03 as indicated. Data represent mean±s.d. of three biological replicates. (B) Western blot analysis of GFP and Tfcp2l1 expression in i-EGFP and i-Tfcp2l1 mESCs cultured in the presence or absence of 0.2 μg/ml Dox for 12 h. (C) i-EGFP and i-Tfcp2l1 mESCs were cultured in N2B27/CHIR for five passages in the presence or absence of Dox. Scale bar, 100 μm. (D, E) Alkaline phosphatase staining of i-Tfcp2l1 mESCs cultured in the presence or absence of Dox and quantification of alkaline phosphatase-positive colonies. Scale bar, 100 μm. Data represent mean±s.d. from triplicate experiments. (F) qRT–PCR analysis of Klf4, Nanog, Cdx2, Fgf5, T, Mixl1, Gata4, and Gata6 expression in i-Tfcp2l1 mESCs cultured in N2B27/CHIR with or without Dox. Data represent mean±s.d. of three biological replicates. (G) Immunostaining for neural marker Nestin, cardiomyocyte marker Myosin, and primitive endoderm marker Gata4 in i-Tfcp2l1 mESC-derived differentiated cells. Hoechst was used for nuclear staining. Scale bar, 100 μm. (H, I) Morphology, alkaline phosphatase staining, and immunostaining of i-Tfcp2l1 mESCs cultured in N2B27/PD03 with or without Dox for five passages. Scale bars, 100 μm.Source data for this figure is available on the online supplementary information page. Source Data for Figure 2B [embj2013175-sup-0002-SourceData-S2.pdf] Download figure Download PowerPoint Forced expression of Tfcp2l1 can maintain self-renewal in the absence of LIF/Stat3 signalling As LIF can be used to replace CHIR or PD03 in the presence of either in maintaining mESC self-renewal (Wray et al, 2010), we sought to determine whether there is any connection between LIF signalling and Tfcp2l1 expression. We evaluated the transcriptional responsiveness of Tfcp2l1 to LIF stimulation in 46C mESCs. 46C mESCs were cultured in the absence of LIF for 12 h, and the expression of Tfcp2l1 as well as the direct Stat3 targets Socs3 and Klf4 (Hall et al, 2009) was then analysed by qRT–PCR, which showed that expression of all three genes was significantly downregulated (Figure 3A). We next cultured the i-Tfcp2l1 mESCs in serum medium in the presence of different concentrations of Dox (Figure 3B). Following LIF withdrawal, i-Tfcp2l1 mESCs remained undifferentiated when Dox was present at a concentration of 0.2 μg/ml or higher (Figure 3B–F), whereas they rapidly differentiated when Dox was absent or present only at a low concentration (0.02 μg/ml) (Figure 3B–E). We then compared Tfcp2l1 expression induced by various concentrations of Dox with endogenous Tfcp2l1 expression induced by LIF and 2i. Because 0.02 μg/ml of Dox induced lower Tfcp2l1 expression than did LIF or 2i (Figure 3D), we chose 0.2 μg/ml of Dox to achieve a more-appropriate level of Tfcp2l1 expression for all the following experiments. Figure 3.Enforced Tfcp2l1 promotes mESC self-renewal independent of LIF/Stat3. (A) qRT–PCR analysis of Socs3, Klf4, and Tfcp2l1 expression in 46C mESCs deprived of LIF for 12 h. Data represent mean±s.d. of three biological replicates. (B) Alkaline phosphatase staining of i-Tfcp2l1 mESCs cultured in the absence or presence of different concentrations of Dox for 8 days. (C) Quantification of alkaline phosphatase-positive colonies. Data represent mean±s.d. from triplicate experiments. (D) qRT–PCR analysis of Tfcp2l1 in i-Tfcp2l1 mESCs cultured in the indicated conditions for 12 h. (E) Morphology of i-EGFP and i-Tfcp2l1 mESCs cultured in serum medium without LIF for 8 days. Scale bar, 100 μm. (F) Immunostaining of i-Tfcp2l1 mESCs after 8 days in the presence or absence of Dox. Scale bar, 100 μm. (G) Western blot analysis of Tfcp2l1 expression in empty or Tfcp2l1 vector-transfected Stat3 knockout ESCs cultured in N2B27/2i. (H) Morphology and alkaline phosphatase staining of Stat3 knockout ESCs transfected with empty or Tfcp2l1 vector and cultured in serum without LIF for one passage. Scale bar, 100 μm. (I) mESCs were deprived of LIF for 12 h, and then treated with indicated kinase inhibitors for 1 h before LIF stimulation for 12 h, as indicated on timeline (lower right). Socs3 and Tfcp2l1 transcripts were evaluated at −1, 1, and 12 h by qRT–PCR. Data represent mean±s.d. of three biological replicates.Source data for this figure is available on the online supplementary information page. Source Data for Figure 3G [embj2013175-sup-0003-SourceData-S3.pdf] Download figure Download PowerPoint To determine whether Stat3 is required for Tfcp2l1-promoted mESC self-renewal, we overexpressed Tfcp2l1 in Stat3-null mESCs (Figure 3G) and cultured them in serum without LIF or 2i (Ying et al, 2008). These cells remained undifferentiated even after multiple passages, whereas Stat3-null mESCs transfected with an empty vector died or differentiated after one passage (Figure 3H). These results indicate that Tfcp2l1 promotes mESC self-renewal independent of Stat3. Binding of LIF to the LIF receptor/gp130 complex can trigger three signalling pathways: janus kinase (JAK)/Stat3, phosphoinositide 3-kinase (PI3K)/AKT, and mitogen-activated protein kinase (MAPK) (Hirai et al, 2011). To determine which of these signalling pathways are involved in the induction of Tfcp2l1 by LIF, we treated 46C mESCs with inhibitors specific for JAK, PI3K, and MAPK, and then assessed the induction of Tfcp2l1 by LIF. Upregulation of Socs3 by LIF stimulation was completely prevented by JAK inhibitor I but not by the PI3K and MAPK inhibitors (Figure 3I). The presence of JAK inhibitor I also prevented the induction of Tfcp2l1 by LIF, whereas PI3K inhibitor had no effect, and treatment with MAPK inhibitor PD03 was associated with strong Tfcp2l1 induction as expected (Figure 3I). Taken together, our results suggest that LIF-induced expression of Tfcp2l1 is mediated by Stat3, but not by PI3K or MAPK. These observations indicate that a high level of Tfcp2l1 is an important factor in liberating mESCs from LIF dependence and in promoting self-renewal downstream of Stat3. Given that CHIR, PD03, and LIF exhibit a similar ability to induce Tfcp2l1 expression, we wanted to examine whether these three mediators depend on one another for their upregulative effect. We pre-treated mESCs with JAK inhibitor I (Stat3 signalling inhibitor), 53AH (Wnt signalling inhibitor) (Willems et al, 2011), or bFGF (activator of FGF/MEK signalling pathway) for 1 h before adding LIF, CHIR, or PD03. Twelve hours after the addition of LIF, CHIR, or PD03, Egr1, a target of the FGF/MEK signalling pathway, was induced by bFGF but repressed by MEK inhibitor PD03 (Figure 4A). Axin2, a target of the Wnt signalling pathway, was induced by CHIR but repressed by 53AH (Figure 4B); and Socs3, a target of the LIF/Stat3 signalling pathway, was induced by LIF but repressed by JAK inhibitor I (Figure 4C). Tfcp2l1 expression was considerably elevated following treatment with LIF, CHIR, or PD03, even in the presence of irrelevant signalling pathway inhibitors/activator (Figure 4A–C), demonstrating that CHIR, PD03, and LIF can induce Tfcp2l1 transcription independent of one another. Figure 4.PD03, CHIR, and LIF induce Tfcp2l1 independently. (A–C) mESCs were deprived of LIF for 12 h, and then treated with 1 μm 53AH, 10 μM JAK inhibitor I, or 20 ng/ml bFGF for 1 h before LIF, CHIR, or PD03 stimulation. Egr1, Axin2, Socs3, and Tfcp2l1 transcript levels were evaluated 12 h later by qRT–PCR. Data represent mean±s.d. of three biological replicates. Download figure Download PowerPoint Downregulation of Tfcp2l1 impairs mESC self-renewal in LIF condition To test whether Tfcp2l1 knockdown has an effect on LIF/Stat3- or 2i-mediated mESC self-renewal, we infected 46C mESCs with lentiviruses encoding two short-hairpin RNAs (shRNAs) specific for Tfcp2l1 mRNA (shTfcp2l1). Stable knockdown (70–80%) of Tfcp2l1 transcript levels was observed following drug selection (Figure 5A). shTfcp2l1 mESCs formed fewer colonies than did mESCs expressing scramble control shRNAs, and these colonies exhibited a comparatively flatter morphology, even in the presence of LIF (Figure 5B and C). Accordingly, these shTfcp2l1 cells expressed a low level of the pluripotency markers Oct4, Sox2, Nanog, and Klf4, but high levels of the differentiation-related genes Cdx2, Nestin, T, Gata6, and FoxA2 (Figure 5D). To exclude off-target effects of the two shRNAs used in Tfcp2l1 knockdown, we modified the Tfcp2l1 expression construct to encode Tfcp2l1 mRNA harbouring synonymous mutations at the two sites targeted by our shRNAs (see Materials and methods), and tested whether transfection with this mutant construct could rescue the knockdown phenotypes. Stable mutant-Tfcp2l1 transfectants formed significantly more alkaline phosphatase-positive colonies in serum/LIF than did empty-vector controls following infection with shTfcp2l1 lentiviruses (Figure 5E and F). These results reveal that Tfcp2l1 can mediate LIF/Stat3-dependent self-renewal in ESCs. shTfcp2l1 mESCs cultured in 2i maintained a higher level of Tfcp2l1 expression than did shTfcp2l1 mESCs in serum+LIF (Figure 5G). Consistent with this, shTfcp2l1 mESCs cultured in 2i maintained an undifferentiated phenotype, although they formed smaller colonies than did mESCs expressing scramble control shRNA (Figure 5H). Figure 5.Knockdown of Tfcp2l1 impairs Stat3- but not 2i-mediated mESC self-renewal. (A) qRT–PCR analysis of Tfcp2l1 expression in shRNA knockdown cells. The transcript level was normalized against scramble shRNA control. Data represent mean±s.d. of three biological replicates. (B) Alkaline phosphatase staining of shTfcp2l1 mESC colonies cultured in serum/LIF condition for 8 days. Scale bar, 100 μm. (C) Quantification of alkaline phosphatase-positive shTfcp2l1 mESC colonies. Data represent mean±s.d. from triplicate experiments. (D) qRT–PCR analysis of ESC pluripotency marker (Oct4, Sox2, Nanog, Klf4, and Tbx3) and differentiation-associated gene (Cdx2, Nestin, T, Gata6, and FoxA2) expression in shTfcp2l1 ESCs cultured in the presence of LIF. Transcript levels were normalized against scramble shRNA control. Data represent mean±s.d. of three biological replicates. (E, F) Alkaline phosphatase staining and quantification of alkaline phosphatase-positive empty vector and mutant-Tfcp2l1 (Tfcp2l1mut) colonies transfected with Tfcp2l1 shRNA lentivirus and cultured in serum/LIF medium. Scale bar, 100 μm. (G) qRT–PCR analysis of Tfcp2l1 expression in Tfcp2l1 shRNA knockdown and scramble shRNA control cells cultured in serum/LIF or N2B27/2i. Data represent mean±s.d. of three biological replicates. (H) Alkaline phosphatase staining of shTfcp2l1 mESC colonies cultured in N2B27/2i for five passages. Scale bar, 100 μm. Download figure Download PowerPoint Tfcp2l1 facilitates epiblast stem cells reprogramming Tfcp2l1 expression, like that of Oct4 and Nanog, is robust in undifferentiated mESCs, but declines abruptly upon induction of embryoid body (EB) formation (Figure 6A), indicating that Tfcp2l1 is a pluripotency marker. On the basis of this finding and previous reports that direct transfection with other Stat3 targets such as Gbx2, Klf4, Klf2, or Nanog can reprogram epiblast stem cells (EpiSCs) to a state of naïve pluripotency (Guo et al, 2009; Guo and Smith, 2010; Yang et al, 2010; Tai and Ying, 2013), we tested whether transfection of EpiSCs with Tfcp2l1 could induce naïve pluripotency. Figure 6.Tfcp2l1 reprograms EpiSCs to naïve pluripotency. (A) 46C mESCs and 46C-derived embryoid bodies were harvested. Oct4, Nanog, and Tfcp2l1 transcript levels were analysed by qRT–PCR. Data represent mean±s.d. of three biological replicates. (B) Morphology and alkaline phosphatase staining of 46C mESCs and 46C mESC-derived EpiSCs cultured in Activin A, bFGF, and 53AH for eight passages. Scale bar represents 100 μm. (C) Expression patterns of Tfcp2l1, Nanog, Rex1, and Fgf5 in 46C mESCs and EpiSCs. Data represent mean±s.d. of three biological replicates. (D) Western blot analysis of Tfcp2l1 in 46C EpiSCs with stable Tfcp2l1 transgene expression. (E) Morphology of empty and Tfcp2l1 vector EpiSC transfectants cultured in serum medium supplemented with LIF/2i for 8 days. Scale bar represents 100 μm. (F) Alkaline phosphatase staining of Tfcp2l1 EpiSCs and three Tfcp2l1 Epi-iPSC (induced pluripotent stem cell) clones. Scale bar represents 100 μm. (G, H) 105 empty and Tfcp2l1 vector-transfected CD1 EpiSCs cultured in serum medium supplemented with LIF/2i. After 10 days, alkaline phosphatase-positive colonies were photographed and counted under a microscope. Scale bar represents 100 μm. (I) Comparison of gene marker expression in Tfcp2l1 Epi-iPS cells and CD1 EpiSCs. Nanog, Klf2, Klf4, Nr0b1, Rex1, and Stella are ESC markers, whereas Fgf5 and Otx2 are EpiSC markers. Data represent mean±s.d. of three biological replicates.Source data for this figure is available on the online supplementary information page. Source Data for Figure 6D [embj2013175-sup-0004-SourceData-S4.pdf] Download figure Download PowerPoint We induced the differentiation of 46C mESCs into EpiSCs, and examined the expression of typical ESC markers. 46C EpiSCs stained negative for alkaline phosphatase and expressed a higher level of Fgf5 and a lower level of Tfcp2l1, Rex1, and Nanog compared with 46C mESCs (Figure 6B and C). 46C EpiSCs were then transfected with a PiggyBac vector encoding Tfcp2l1 and analysed by western blotting for expression of Tfcp2l1 (Figure 6D). Empty-vector control 46C EpiSCs died or differentiated within 8 days of being switched to serum/LIF/2i, whereas Tfcp2l1-46C EpiSCs gave rise to many ESC-like colonies with dome-shaped morphology (Figure 6E). We picked up three of these colonies and transferred them to normal mESC culture medium (serum/LIF without 2i). After two passages, these cell populations retained robust alkaline phosphatase activity (Figure 6F). To address the possibility that these results reflect mixed populations of reprogrammed 46C cells and original 46C mESCs, we derived EpiSCs from embryonic day 5.5 CD1 mouse embryos, and then introduced Tfcp2l1. One million transfected EpiSCs were cultured for 10 days in serum/LIF/2i and gave rise to ∼80 alkaline phosphatase-positive ESC-like colonies (Figure 6G and H). These converted ESC-like cells could be continuously expanded in serum/LIF without feeders, and expressed high levels of the naïve pluripotency markers Nanog, Rex1, Nrob1, Stella, Klf2, and Klf4, but low levels of the EpiSC-specific markers Fgf5 and Otx2 (Figure 6I). In contrast, far fewer colonies arose from empty-vector transfectants and these colonies died or differentiated after passaging. These results show that Tfcp2l1 is capable of converting EpiSCs into a state of naïve pluripotency. Tfcp2l1 contributes to ground-state pluripotency via Nanog The expression of pluripotency genes Oct4, Sox2, Klf4, and Nanog was downregulated in shTfcp2l1 mESCs (Figure 5D). We examined the induction