Abstract
Summary
Maintenance of pluripotency is a multifactorial process in which NF-κB is a negative regulator. Our previous work identified a chromatin role for IκBα, the master regulator of NF-κB signaling, that is critical for the proper regulation of various tissue stem cells. Here, we found that IκBα accumulates specifically in the chromatin fraction of pluripotent embryonic stem cells. IκBα depletion does not affect NF-kB-dependent transcription, but causes a profound epigenetic rewiring in pluripotent stem cells, including alterations in H3K27me3, a histone mark catalyzed by Polycomb repression complex 2. Chromatin changes induced by IκBα depletion affect a subset of pluripotency genes and are associated with altered gene transcription. At the cellular level, IκBα-deficient embryonic stem cells are arrested in a naive pluripotency state when cultured in serum/LIF conditions and fail to exit pluripotency under differentiation conditions. By constructing separation-of-function mutants, we show that the effects of IκBα in regulating stem cell pluripotency are NF-κB-independent, but mainly rely on its chromatin-related function. Taken together, our results reveal a novel mechanism by which IκBα participates in the regulation of the pluripotent state of embryonic stem cells and shed light on the interplay between inflammatory signals and the regulation of pluripotency.
Introduction
Embryonic Stem Cells (ESCs) pluripotency is characterized by the capacity to generate all somatic and germline lineages both in vitro and in vivo, depending on the culture conditions. When cultured in serum plus Leukemia Inhibitory Factor (LIF) (Serum/LIF), murine ESCs (mESCs) include cells interconverting to different metastable states 1 that either resemble preimplantation blastocyst (naïve pluripotency) or post-implantation (primed pluripotency) embryonic stages 2–5. The heterogeneity of mESCs in Serum/LIF is reduced by culturing them in the presence of GSK3β and MEK1/2 inhibitors (CHIR99021 and PD0325901, respectively) along with LIF (2i/LIF). Under these conditions, mESCs enter a ground state of naïve pluripotency characterized by self-renewal activity while suppressing any pro-differentiating signals 6,7. This pluripotent state closely resembles the inner cell mass of the E4.0 mouse preimplantation blastocyst 8. After implantation, pluripotent capability of the mouse embryo (E5.5) becomes restricted to Epiblast Stem Cells (EpiSCs), which represent a primed pluripotent state, characterized by coexpression of pluripotency and lineage specification markers 9,10. EpiSCs can give rise to cells from the three germ layers (endoderm, mesoderm and ectoderm), albeit their contribution to chimeras or to germ cells is greatly compromised 4,11. EpiSCs can be derived in vitro from naïve pluripotent cells when cultured in a medium supplemented with Nodal and fibroblast growth factor (FGF) 12. Different epigenetic features distinguish naïve from primed pluripotent states 13, such as changes in the X chromosome inactivation in female cells 12,14, histone post-translational modifications 10 or a global DNA hypomethylation profile observed in naïve pluripotent stem cells 15–17. These epigenetic changes are a prerequisite for the subsequent activation of gene circuits associated with pluripotency exit and germ layer specification 9,10. Crucial regulatory elements for the transition from naïve to primed pluripotency state are located in distal DNA regions or enhancers to facilitate the resolution toward the primed state 18,19. Therefore, identifying novel players to fine-tune the equilibrium between ground and primed states is crucial for preserving both pluripotency stability and differentiation capability in mESCs.
There is now strong evidence that inflammatory signals are critical for stem cell development 20–22, with NF-κB being the main effector. Different strategies to inhibit or attenuate NF-κB in ESCs lead to increase pluripotency gene expression and impair cell differentiation 23,24, or it facilitates reprogramming to induced pluripotent stem cells (iPSC) 25. Canonical NF-κB signaling is triggered by the IKK kinase complex, which induce the phosphorylation and subsequent degradation of the NF-κB inhibitor, IκBα, leading to nuclear translocation of the NF-κB factors (e.g. p50/p65) 26. However, we have previously identified a nuclear IκBα function, which is critical for tissue stem cell homeostasis and the proper differentiation of epidermal, intestinal and hematopoietic stem cells 27–30. IκBα chromatin function is mediated by the interaction with histone deacetylases (HDACs), core elements of the Polycomb Repressor Complex 2 (PRC2) and histone H2A and H4 27,29,31.
We have now investigated the role of IκBα in the context of pluripotent stem cells. Whereas cytoplasmic IκBα is detected in the different states and its levels decrease after differentiation, in agreement with NF-κB activity as a promoter of differentiation, we specifically detected chromatin-bound IκBα in the naïve pluripotent stem cells. Importantly, IκBα depletion in ESCs did not affect NF-κB activity but it stabilizes the naïve (2i/LIF-like) state of pluripotency under Serum/LIF conditions, displaying an epigenetic rewiring, including changes in the PRC2-dependent H3K27me3 histone mark and impacting in proximal and distal regulation of pluripotent genes. Using a new Separation-Of-Function (SOF) IκBα mutants32, we have now definitively demonstrated that chromatin-bound IκBα is required for naïve pluripotency exit of mESCs in an NF-κB-independent manner. Overall, we identified IκBα as a novel key player in the regulation of the exit from naïve-to-primed pluripotency and essential for the activation of gene programs involved in germ layer specification.
Results
IκBα and NF-κB have opposite expression dynamics in naïve pluripotent cells
Due to the prominent role of NF-κB signaling in mESCs regulation, we analyzed the expression pattern of the different NF-κB members including the IκB inhibitors in the three defined pluripotent stages: ground state of naïve pluripotency (2i/LIF), naïve pluripotency (Serum/LIF), and primed pluripotency (Epiblast Stem Cells or EpiSCs) 5 (Figure 1A). In agreement with NF-κB being a pro-differentiation factor, canonical NF-κB inhibitors including IκBα (Nfkbia), IκBβ (Nfkbib) and IκBε (Nfkbie) were all expressed in the naïve pluripotency state (both 2i/LIF and Serum/LIF), with Nfkbia levels gradually reduced upon naïve pluripotency exit towards EpiSCs (Figure 1B). In contrast, Rela, Nfkb1 and Nfkb2 genes, codifying for the NF-κB factors p65/RelA, p105 and p100, respectively, showed opposite expression dynamics increasing the levels in EpiSCs (Figure 1C). We further characterized the distribution of IκBα in the cytoplasm, nucleus and chromatin fractions of the three states of pluripotency (Figure 1D). Total IκBα protein levels correlated with the detected RNA levels in the three different cellular states being highest in the 2i/LIF condition (Figure 1B), with a large accumulation in the cytoplasm. A fraction of IκBα protein was detected in the chromatin of naïve pluripotent cells, being highest in cells cultured in Serum/LIF conditions compared to 2i/LIF and absent in the EpiSCs. (Figure 1D). The dynamics of IκBα RNA and protein levels from naïve pluripotent ESCs to EpiSCs was further corroborated by analysis of an additional database that includes both naïve and primed pluripotent states33 (Figure 1E).
Next, we studied IκBα expression in differentiated embryoid bodies (EBs) from mESCs (Serum/LIF) (see materials and methods) and analyzed the expression levels of different pluripotency, differentiation-linked, and NF-κB genes by RNA sequencing. Nfkbia (IκBα gene) was expressed at higher levels specifically in mESCs (Serum/LIF) whereas cells undergoing differentiation (48h and 96h) displayed a reduction in its expression levels (Figure 1F). The expression pattern of Nfkbia was similar to the pluripotency genes whereas most of the NF-κB genes were upregulated upon differentiation, clustering together with developmental genes, as previously described 24. Notably, several polycomb elements were highly expressed in mESCs, consistent with the possibility that chromatin-bound IκBα modulates PRC2 activity 27 in the pluripotent state.
Taken together, these data show that, unlike the other NF-kB inhibitors and target genes, the expression of Nfkbia is high in the naive pluripotent state, with IκBα protein present in the cytoplasm and chromatin of naive pluripotent cells. Moreover, whereas canonical NF-κB subunits are primarily absent, we detected several Polycomb elements expressed in mESCs.
IκBα is required in mESCs to exit the naïve pluripotency state
To investigate the functional implication of IκBα in naïve pluripotency, we knocked-out (KO) IκBα protein in mESCs by CRISPR-Cas9 (IκBα-KO mESCs) (Figure S1A and materials and methods). Notably, IκBα KO mESCs have an impaired ability to undergo differentiation and cannot properly switch off the pluripotency program after 216h (9 days) of pro-differentiation signals, as it is demonstrated by high levels of pluripotency markers OCT3/4 and NANOG (Fig 1A and Fig S1C), higher number of alkaline phosphatase cells (Fig 1B and Fig S1) and higher percentage of SSEA-1+ cells (Fig S1B) within the 216h EBs. These results were also supported by the detection of increased expression level of the naïve pluripotency genes Pou5f1 (Oct3/4 gene), Gbx2, Klf2, Sox2, Zfp42 (Rex1) and Nanog in IκBα KO EBs after 216h of differentiation (Figure S1B).
To shed light into the molecular mechanisms governing the differentiation impairment in IκBα KO EBs, we performed RNA-seq of IκBα WT and IκBα KO mESCs in the first stages of the embryoid body differentiation. Analysis of the data indicated that IκBα KO EBs cannot successfully evolve towards differentiation trajectories, as shown by principal component analysis (PCA) of IκBα-WT and IκBα-KO EBs at 48h and 96h in differentiation media (Figure 2C). Analysis of naïve pluripotency markers at 96h (Gab1, Sox2, Esrrb, Zfp42, Dppa4, Klf2, Prdm14, Klf4, Tfcp2l1) also showed higher expression levels in IκBα KO compared to their IκBα WT counterparts (Figures 2D and S1F). Defective differentiation capacity of IκBα KO EBs involved programs associated with specification of all 3 germ layers (endoderm, mesoderm, and ectoderm) (Figures 2E-G). Moreover, IκBα KO EBs were smaller in size (Figure S1E), in agreement with the reduced developmental potential of IκBα-depleted mESCs 34. To further study the requirement of IκBα in pluripotency exit in vivo, we established teratomas by subcutaneous injection of IκBα-WT and IκBα-KO mESCs (Figure 2H and materials and methods) into NSG (NOD.Cg-Prkdcˢᶜⁱᵈ Il2rgᵗᵐ¹ᵂʲˡ/SzJ) mice. IκBα-KO teratomas showed a decreased differentiation potential, as indicated by higher number of OCT4+ cells 6 weeks after injection (Figures 2H-J). These observations further confirm the essential role of IκBα in the exit from the pluripotency state not only in vitro but also under in vivo differentiation signals. To understand the conditions in which IκBα is required for pluripotency exit, we differentiated IκBα-WT and IκBα-KO mESCs cultured in Serum/LIF (naïve pluripotency) towards EpiSCs (primed pluripotency) (Figure 3A, upper panel). IκBα-KO cells were not committed towards the primed pluripotency stage and maintained an elevated expression levels of naïve pluripotency genes (Dppa3, Nanog, Sox2, Rex1, Klf2, Klf4, Gbx2, Tbx3) (Figure 3A, bottom panel). Additionally, IκBα-KO cells retained a naïve pluripotent morphology, forming tight clusters that resemble cells cultured in 2i/LIF medium (Figure S2A).
mESCs cultured in serum/LIF are highly heterogeneous, comprising a mixture of cell states that resemble developmental transitions from preimplantation (naïve pluripotency) to postimplantation (primed pluripotency) embryo 2,3. To investigate how IκBα deficiency was affecting the exit of mESCs from naïve pluripotency, we performed a Gene Set Enrichment Analysis (GSEA) using RNA-seq data from IκBα-KO and IκBα-WT mESCs cultured in Serum/LIF against gene signatures for 2i/LIF and Serum/LIF pluripotency 35. GSEA results revealed a significant enrichment in Serum/LIF IκBα-KO mESCs transcriptome for genes specifically expressed in the naïve ground state (2i/LIF), while they negatively correlate with mESCs cultured in Serum/LIF (Figure 3B) 35. These results agree with the fact that IκBα-KO mESCs cultured in Serum/LIF resemble 2i/LIF morphology, with homogeneous tight clusters of cells, whereas IκBα-WT mESCs retain the colony heterogeneity (Figure S2B). Interestingly, the higher levels of ground-state-related genes Zfp42 (Rex1), Klf2 and Tbx3 in IκBα-KO mESCs were partially obtained when IκBα-WT mESCs were cultured in 2i/LIF for two consecutive passages (Figure 3C).
One of the key features of the naïve pluripotency state is its global DNA hypomethylation pattern 15,16. We assessed the DNA methylation status of IκBα-WT and IκBα-KO mESCs cultured in Serum/LIF conditions by analysis of 5-methyl-cytosine (5mC) mark and using DNA methylation arrays 36. We found that IκBα-KO mESCs contained much lower levels of 5mC compared to IκBα-WT cells (Figure 3D). Moreover, DNA methylation arrays confirmed a global pattern of DNA hypomethylation in IκBα-KO mESCs, which was maintained in 96h EBs (Figure 3E). Interestingly, the DNA hypomethylation status occurred in a genome-wide fashion affecting all chromosomes (Figure S2C), in accordance with a higher pluripotent state of IκBα-KO mESCs.
Overall, these data demonstrate that IκBα is required for the exit of the naïve state of pluripotency and its deficiency results in impaired differentiation of mESCs in vivo and in vitro. Accordingly, IκBα-KO mESCs cultured in Serum/LIF showed genome-wide pattern of DNA hypomethylation.
Lack of IκBα causes an epigenetic rewiring in pluripotent stem cells to resemble 2i/LIF naïve pluripotency in Serum/LIF culture
ChIP Enrichment Analysis (ChEA) of genes identified in our RNA-seq data of IκBα-KO and WT mESCs revealed that differentially expressed genes were putative targets of chromatin regulators including the PRC2 subunit SUZ12 and MTF2 (Figure 4A). We previously described that IκBα is important for the proper deposition of the H3K27me3 mark in different types of tissue stem cells by associating with elements of the PRC2 complex 27,28. Since 2i/LIF and Serum/LIF states are characterized by important differences in epigenetic marks, we carried out an epigenetic profiling of IκBα-WT and IκBα-KO mESCs cultured in Serum/LIF. We performed chromatin immunoprecipitation followed by sequencing (ChIP-seq) of histone modifications associated with gene activation (H3K4me3) and gene repression (H3K27me3) (Figures 4B and S3A-B). Overall, Serum/LIF IκBα-KO mESCs exhibited a general increased H3K4me3 status and redistribution of the PRC2-catalyzed H3K27me3 mark (Figure 4B). Notably, gain of the H3K4me3 mark was associated with a reduction of H3K27me3 levels in genes associated with naïve pluripotency function (Figures 4B-C and S3A-B). In particular, 41 genes with concomitant gain of H3K4me3 and loss of H3K27me3 were found to be enriched in mechanisms associated with pluripotency function (Figure 4C), including Tbx3 and Gbx2 (Figure 4D-4E). A second set of pluripotency-related genes showed an increase in H3K4me3 without changes in H3K27me3 (Zfp42 or Klf2). Both sets of pluripotency genes showed increased expression in the absence of IκBα (Figure 4H).
We next aimed to study whether the lack of IκBα was also affecting distal regulatory regions by analyzing enhancer-associated histone marks in IκBα-WT and IκBα-KO mESCs. We quantified the total amount of H3K27Ac (active enhancer) and H3K4me1 (poised enhancer) by ChIP-seq experiments conducted in both IκBα-WT and IκBα-KO mESCs. Overall, IκBα-KO mESCs showed a reduction in poised enhancers at genes (or related genomic regions) associated with differentiation processes (Figures 4F and S3C) and an increase in active enhancers at genes associated with pluripotency (Figure 4F and S3D), such as Tbx3, Tfcp2l1 and Zfp42 (Rex1) (Figure 4G), which positively favors their higher expression in IκBα-KO mESCs (Figure 4H). These results further support that mESCs lacking IκBα are epigenetically remodeled to favor the ground state of naive pluripotency, which may negatively impact on their differentiation potential, similar to that described for other models with stabilized naïve pluripotent state37,38.
The chromatin function of IκBα in pluripotency exit is independent of classical NF-κB activity
Although previous results from C. elegans 39 and Drosophila 27 support the functional relevance of chromatin-related IκBα function, the investigation of this alternative IκBα activity remains challenging due to the predominant role of this protein in NF-κB regulation. However, and similar to that found in other models, we did not detect major changes in canonical NF-κB target genes upon IκBα depletion neither in Serum/LIF mESCs, nor 48h and 96h EB-differentiating conditions (Figure 5A). Thus, either NF-κB regulation does not require IκBα at this stage or it might be compensated by the other IκBs (IκBβ, IκBε or p100), as previously demonstrated 40,41. This observation suggests that either NF-κB signaling is not active in naïve pluripotency, which is supported by the low expression levels of NF-κB subunits (see Figures 1B-C and 1E-F), or IκBα function on NF-κB is compensated by the other IκBs (IκBβ, IκBε or p100), as previously demonstrated40,41.
To further investigate the relative impact of both IκBα functions in mESCs, we took advantage of recent results from our group that led to the identification of the specific IκBα protein residues that define NF-κB- or chromatin-binding through H2A/H4. Mutation of these residues allowed the generation of doxycycline-inducible (i) separation-of-function IκBα mutants that are specifically deficient in one or the other function, at least in the intestinal epithelial cells32. We called i-IκBαΔChromatin the i-SOF IκBα mutant that interacts with NF-κB but is deficient for H2A and H4 binding and i-IκBαΔNF-κB the one that interact with histones but is deficient for NF-κB subunits binding (Figure 5B and 32). The three different IκBα forms (i-IκBαWT, i-IκBαΔNF-κB and i-IκBαΔChromatin) were stably transfected in the IκBα KO mESCs and induced by 16h of doxycycline treatment (Figure S4A). We used these cells to investigate the biochemical properties and functional impact of SOF IκBα mutants in mESCs. By pull-down experiments, we found that i-IκBαΔNF-κB expressed in ESCs was able to interact with histone H2A but not with the NF-κB member p50. In contrast, i-IκBαΔChromatin interacts with p50 but not with H2A, while i-IκBαWT preserves both binding capacities (Figure 5C). In concordance with these results, i-IκBαWT and i-IκBαΔNF-κB were found in both the cytoplasmic and chromatin fractions of mESCs, whereas IκBαΔCromatin was retained in the cytoplasm (Figures 5D-E). In multiple experiments, we noticed a significant reduction in protein levels of IκBαΔchromatin despite the mRNA expression levels of the different IκBα forms being comparable (Figures 5C-D and S4A).
Then, we studied whether i-IκBαWT and/or the i-SOF IκBα mutants (IκBαΔNF-κB or IκBαΔChromatin) were able to reverse any of the phenotypes observed in the IκBα-KO mESCs (Figure S4B). Induction of i-IκBαWT and i-IκBαΔNF-κB reduced the expression levels of the naïve pluripotent genes Zfp42, Klf2, Sox2 and Tbx3, which were increased by i-IκBαΔChromatin (Figure 5F). In addition, the ground state-associated DNA hypomethylation was specifically reverted in i-IκBαWT and i-IκBαΔNF-κB mESCs but not in i-IκBαΔChromatin mESCs (Figure 5G). Finally, we addressed whether i-IκBαWT, i-IκBαΔNF-κB and i-IκBαΔChromatin mESCs were capable to revert the differentiation blockage of IκBα-KO ESCs (Figure S4B). Induction of IκBαWT and IκBαΔNF-κB was enough to restore the differentiation potential of IκBα-KO mESCs into EBs, as indicated the increase in the number of differentiated cells (based on percentage of SSEA-1neg cells) (Figures 5H and S4C) and the reduced number of AP+ colonies (in the 216h IκBα-KO reconstituted EB assays), which was similar to the number of colonies obtained from IκBαWT EBs (Figures 5I). Lastly, the number of remaining pluripotent-like cells (defined by OCT3/4 NANOG staining) was also reduced in i-IκBαWT and i-IκBαΔNF-κB 216h EBs, and only small and few regions of undifferentiated cells was observed. In contrast, induction of i-IκBαΔChromatin did not restore the differentiation potential of IκBα-KO mESCs (Figures 5J and S4D).
These results demonstrate that the chromatin-dependent IκBα function, but not its NF-κB-related activity, is necessary for the proper regulation of chromatin marks at specific genomic regions that are linked to transcriptional changes that are likely at the base of the capacity of ESCs to exit the ground state of naïve pluripotency and differentiate.
Discussion
Here we have identified a new and unexpected function for the IκBα protein in the regulation of the naïve pluripotency exit. Using mESCs to investigate the role of IκBα in pluripotency maintenance and differentiation, we discovered that the absence of IκBα severely impairs the exit of pluripotency. Furthermore, IκBα-KO mESCs stabilizes the ground state of naïve pluripotency under Serum/LIF conditions, shedding light on its function as a critical regulator of the transition from naïve to primed pluripotency. While IκBα is commonly known for its role as an inhibitor of the NF-κB signaling pathway 26, our laboratory has previously furnished compelling evidence of its alternative role through its interaction with histones and other chromatin components 27,29,31. We have demonstrated that chromatin-bound IκBα plays a crucial role in regulating skin and intestinal stem cells 27,28, hematopoietic stem cell development 42, and it influences the regenerative capacity of these tissues, and in some cases, their susceptibility to neoplastic transformation. Disentangling the chromatin-related function of IκBα from its canonical role as an inhibitor of NF-κB has been challenging, making it difficult to determine its truly biological significance. To tackle this challenge, we engineered separation-of-function (SOF) mutants of IκBα, as detailed in 43, by identifying specific residues that are essential for binding either NF-κB elements or histones. Interestingly, we have shown that only the histone-binding proficient IκBα mutant (IκBαΔNF-κB), which lacks the ability to bind to NF-κB, has the capability to reverse the stabilization of the ground state observed in IκBα-KO mESCs. This is sufficient to facilitate their exit from the state of naïve pluripotency, restoring their full differentiation potential (Figure 5). The involvement of NF-κB in pluripotency has been a subject of prior investigation. Studies have revealed that pluripotent cells display a dampened inflammatory signaling pathway, which is induced upon pluripotency dissolution and activation of differentiation 24,44. However, the precise molecular mechanisms underlying the attenuated status of NF-κB during this stage remain incompletely understood. A recent work has shed light on a potential regulatory axis involving ATG5-β-TrCP1-NF-κB, proposing it as a mechanism that diminishes NF-κB activity by stabilizing the IκBα protein in mESCs 23. In any case, since modification of canonical NF-κB activity directly impacts on the levels IκBα expression, the previous NF-κB-associated phenotypes should be reevaluated considering this chromatin role of IκBα. Furthermore, the expression of IκBαΔChromatin, which retains the inhibitory function of NF-κB, failed to mitigate the ground state stabilization observed in IκBα-KO mESCs (Figure 5). This finding suggests that the observed role of IκBα in regulating the exit from naïve pluripotency is not due to NF-κB inhibition. In fact, there is evidence that the chromatin function of IκBα is an ancestral function preceding its role as NF-κB inhibitor. In Caenorhabditis Elegans (C.elegans), which lacks NF-κB orthologs but possesses two homologs of IκBα, the absence of IκBα orthologs has severe multi-organ differentiation defects 39. This observation further supports the notion that the chromatin-related function of IκBα plays a fundamental role in developmental processes, independent of its canonical role in NF-κB signaling.
We have observed that IκBα-KO mESCs exhibit epigenetic and transcriptomic profiles resembling the ground state of naïve pluripotency under Serum/LIF culture (Figure 3). In this regard, the fact that IκBα is preferentially located in the chromatin of mESCs cultured in Serum/LIF, when they exhibit a high degree of heterogeneity, suggests that IκBα might act as a regulator of this heterogeneity. One potential explanation might be that IκBα could be affecting the stability of fluctuating transcription factors, which is crucial to mediate the pluripotent-to-differentiation balance 3. Thus, the lack of IκBα might stabilize those transcription factors that would favor the ground-state observed in IκBα-KO mESCs cultured in Serum/LIF. Further experiments are required to demonstrate this hypothesis. Moreover, the differences in murine and human pluripotency 11 point out the necessity to address the real impact of IκBα modulation in the stabilization of the ground state in human pluripotent stem cells.
In summary, our work establishes that IκBα mediates the exit from naïve pluripotency by modulating the activation of naïve pluripotency genes. Importantly, this newly identified role of IκBα operates independently of its canonical function in inhibiting the NF-κB pathway. Our findings underscore the intricate interplay between inflammation and pluripotency status, revealing a previously unrecognized complexity in their interaction.
Methods
Cell culture
mESC line ES-E14TG2a (ATCC; Cat #CRL-1821) was cultured on plastic dishes precoated with 0.1% (w/v) Gelatin (Sigma; Cat #G2500-100G) in Serum/LIF medium. Serum/LIF is composed by DMEM basal medium (Sigma-Aldrich; Cat #D5796) supplemented with 15% FBS (ESC-qualified; Gibco Cat #26140079), 1X Glutamax (Gibco; Cat #35050061), 1X NEAA (Gibco; Cat#11140050), 1mM Sodium Pyruvate (Gibco; Cat#11360070), 1000U/mL Leukemia Inhibitory Factor (LIF) (Millipore, Cat #ESG1107) and 0.125mM 2-mercaptoethanol (Gibco; Cat#31350010). Medium was changed every day, and cells were splitted every other day using TrypLE Express (Gibco; Cat #12605010) for harvesting. Cells were maintained in a 5% CO2 incubator at 37°C.
For inducing the ground-state of naïve pluripotency, mESCs were cultured in 2i/LIF medium for 2 consecutive passages. 2i/LIF medium is composed by NDiff 227 medium (TAKARA, Cat #Y40002) supplemented with 1000U/mL LIF, 0.4 µM PD032591 (Selleck Chemicals, Cat #S1036) and 3 µM CHIR99021 (Merck, Cat #SML1046).
Epiblast stem cell differentiation
Differentiation from mESCs towards Epiblast Stem Cells (EpiSCs) was performed as previously described 45. 3x104 mESCs were seeded in 6-well plastic dishes, and they were cultured in Serum/LIF medium for 24h. Medium was then switched to NDiff 227 medium (TAKARA, Cat #Y40002) supplemented with 20 ng/ml activin A (Cat. #120-14E; PeproTech) and 10 ng/ml Fgf basic (R&D Systems, Cat #233-FB-025/CF). Cells were submitted to daily media changes till day 5 (120h), when they were further analyzed.
Embryoid bodies differentiation from mESCs
Embryoid bodies differentiation was established as described in 46. Briefly, mESCs were splitted twice in Serum/LIF medium before inducing differentiation. Once mESCs are 80% confluent, cells were collected using TrypLE Express. One well of 6-well plate was splitted in an entire 6-well plate in IMDM-ES medium for 48h. IMDM-ES medium is composed of Iscove’s Modified Dulbecco’s Medium (IMDM) (Cytiva, Cat #16SH30259.01) supplemented with 20% FBS (ESC-qualified; Gibco Cat #26140079), 1X Glutamax (Gibco; Cat #35050061), 1X NEAA (Gibco; Cat#11140050), 1mM Sodium Pyruvate (Gibco; Cat#11360070), 10ng/mL Leukemia Inhibitory Factor (LIF) (Millipore, Cat #ESG1107) and 0.125mM 2-mercaptoethanol (Gibco; Cat#31350010). For embryoid body induction, mESCs were harvested, and they were rinsed twice with DPBS (Gibco; Cat #14190144). Cells were very well disaggregated into single-cells, and 1.2 x 104 cells/mL embryoid body differentiation (EBdiff) medium were resuspended to a total volume of 25mL of EBdiff medium. EBdiff medium is composed by IMDM supplemented with 15% FBS (ESC-qualified; Gibco Cat #26140079), 1X Glutamax (Gibco; Cat #35050061), 50 μg/mL ascorbic acid (Sigma; Cat #A-4544), 180 μg/mL Transferrin (Roche; Cat #10652202001) and 0.45 mM alpha-monothioglycerol (MTG) (Sigma; Cat #M6145). EBs were formed in suspension for 5 days (120h). At day 5, in order to elongate the differentiation up to 216h (day 9), EBdiff medium was refreshed by harvesting EBs with 10mL serological pipette, centrifuge at 200 g for 3 min and EBs were resuspended in fresh EBdiff medium for 4 more days.
CRISPR/Cas9 gene editing and cell lines generation in mESCs
The two guide RNAs (gRNAs) targeting the Nfkbia locus were designed using the CRISPR design tool from MIT (http://crispr.mit.edu). The best 2 gRNAs (based on on-target and off-target scores) that were targeting the exon 1 of Nfkbia (IκBα gene) were selected. After annealing, one of the gRNAs was cloned into SpCas9(BB)-2A-GFP (px458) plasmid (Addgene; Cat #48138), and the other gRNA was cloned into the px330-mCherry plasmid (modified from Addgene; Cat #98750 to incorporate mCherry reporter). mESCs were co-transfected with the two plasmids (px458-gRNA1 and px330-mCherry-gRNA2). 3x105 cells were seeded a day before per well of 6-well plate. At day of transfection, cells were washed once with 1X DPBS (Gibco; Cat #14190144), and 2mL Opti-MEM medium (Gibco; Cat #31985070) was added into cells. 1.25 μg of each plasmid (2.5 μg of total DNA) was incubated with 10 μL of Lipofectamine 2000 (Invitrogen; Cat #11668019) in 240 μL of Opti-MEM for 20 min. OptiMEM::Lipofectamine::DNA mixture was added into mESCs, and cells were incubated for 5 hours. Cells were then washed with DPBS, and medium was replaced by Serum/LIF. 48h after transfection, GFP+ mCherry+ single cells were sorted by FACS using BD FACSAria II Cell Sorter (BD Bioscience). Mutant clones were screened through PCR and western blotting to identify single clones with no IκBα protein expression. Non-targeting scrambled gRNAs were cloned into px458 and px330-mCherry plasmids as wildtype clones. Three independent clones from each genotype (IκBα-WT and IκBα-KO) were selected for further experiments.
Doxycycline-inducible IκBα (IκBαWT, IκBαΔNF-κB and i-IκBαΔH2A/H4) mESCs were generated by cloning the three different IκBα versions into a PiggyBac transposon system 47. IκBα protein-coding cDNAs were PCR-amplified, and NheI and SalI restriction sites were placed at the 5’ and 3’ ends, respectively. T2A-EGFP fragment was amplified from PX458 plasmid, and SalI and AgeI restriction sites were introduced at its 5’ and 3’ ends. PB-TRE backbone (Addgene, Cat# 63800) was digested with NheI and AgeI enzymes, and mNfkbia and T2A-EGFP fragments were ligated into the PB-TRE digested vector. To generate mESCs containing the inducible vector, IκBα-KO mESCs were co-transfected with PB-TRE-mNfkbia-T2A-EGFP and PiggyBac transposase plasmids, and pool of transfected cells was selected by Hygromycin (100µg/mL) for 7 days. Cells were doubly screened by FACS sorting green fluorescent protein after 24h of doxycycline (1µg/mL) treatment. Pool of cells were used for further experiments. Oligos sequences are included in supplementary table 1.
Teratoma formation assay
IκBα-WT and IκBα-KO mESCs grown in Serum/LIF (5 × 105) were injected intramuscularly into severe combined immunodeficient mice (NSG). In order to favor 3D aggregation of cells and teratoma formation, cells were resuspended in Matrigel Matrix (BD; Cat #356234) prior to injection. Six weeks later, mice with tumors were euthanized, and tumors were fixed in formaldehyde, embedded in paraffin, sectioned and stained with hematoxylin and eosin for histological analysis.
Immunofluorescence staining
For immunostaining of mESCs, cells were seeded on 0.1% gelatin-coated coverslips on 6-well plates; for EB immunofluorescence, they were collected directly from the plate. Samples were washed twice with 1X PBS, and they were fixed at 4°C for 30 minutes with 4% PFA (Electron Microscopy Sciences; Cat #15713S). For immunofluorescence of 5-Methylcytosine (5mC), fixed cells were incubated with 2M Hydrochloric Acid (Sigma-Aldrich, Cat #H1758) for 20 minutes at room temperature. Samples were then washed twice with 1X Tris-buffered saline (TBS; 50 mM Tris-Cl, pH 8) and permeabilized and blocked with 1X TBS supplemented with 1% Triton-X100 (MERCK; Cat #9036-19-5) and 6% FBS (Biological Industries; Cat #04-001-1A) for 2h at 4°C. Samples were washed twice with 1X TBS supplemented with 6% FBS. Primary antibody incubation was performed in 1X TBS plus 6% FBS and 0.3% or 0.5% Triton-X100 (0.3% for 2D culture and 0.5% for EBs) overnight at 4°C. The following primary antibodies were used: OCT3/4 (1:250; Santa Cruz; Cat #sc-5279), NANOG (1:250; Novus Biologicals; Cat #NB100-588), 5-Methylcytosine (1:500, Invitrogen, Cat #MA5-24694), H3K27Ac (1:2000, Abcam, Cat #AB4729). Samples were then washed 4 times with 1X TBS, 5 min each washing. Secondary antibody incubation was performed in TBS 1X plus 1% BSA (Sigma-Aldrich; Cat #9048-46-8) for 2h at room temperature. The following antibodies were used: Alexa Fluor 488 donkey anti-mouse antibody (1:1000; Invitrogen; Cat #A-21202), Alexa Fluor 647 donkey anti-mouse antibody (1:1000; Invitrogen; Cat #A-31571), Alexa Fluor 594 donkey anti-rabbit antibody (1:1000; Life Technologies; Cat #A-21207), Alexa Fluor 647 donkey anti-rabbit antibody(1:1000, Invitrogen; Cat #A-31573). Samples were washed three times with 1X TBS for 5 min at room temperature each rinse. Samples were mounted using DAPI Fluoromount-G (Southern Biotech; Cat #0100-20).
Microscopy and image acquisition
Fluorescence images were acquired using Confocal Leica TCS SP5 (Leica Microsystems), and Leica application software LAS AF (Leica Microsystems) was used to visualize the images. Images of teratoma haematoxylin and eosin and immunohistochemistry stainings were acquired using BX61 Olympus Microscope (Olympus), and PRECiV™ 2D Image and Measurement Software (Olympus) was used to visualize the images. The Image J (version 1.15) was used for further analysis 48.
Flow cytometry sample preparation and analysis
Cells were collected and they were purified based on FACS using BD FACSAria II Cell Sorter (BD Bioscience). Cells were sorted at 3,500 events/second, and at maximum flow rate of 4 and 85 μm nozzle. 5 μg/ml of DAPI (Biotium; Cat #BT-40043) was used as a viability dye. For flow cytometry analysis of pluripotency exit, cells were disaggregated using TrypLE Express, and they were incubated at 4°C for 20 minutes with SSEA1-eFluor 660 Monoclonal Antibody (1:200, Invitrogene, Cat #50-8813-42). BD LSRFortessa Cell Analyzer or BD LSR II Flow Cytometer (BD Bioscience) were used for Flow Cytometry Analysis experiments. Flow cytometry data were analyzed using FlowJo X v10.0.7 (BD Biosciences).
RNA isolation, cDNA synthesis and quantitative RT-PCR
Total RNA isolation from cells was performed using the RNeasy Plus Mini Kit (Qiagen; Cat #74136) or RNeasy Micro Kit (Qiagen; Cat #74004) following manufacturer’s instructions. Amount of RNA was quantified with Nanodrop (Thermo Fisher; Cat #ND2000CLAPTOP), and 2 μg of total RNA was retro-transcribed using Transcriptor First Strand cDNA Synthesis Kit (Roche; Cat #04897030001) following the manufacturer’s instructions.
Quantitative RT-PCR was performed in triplicates for each sample, and SYBR Green I Master Kit (Roche; Cat #04887352001) was used to carry out the reaction. qRT-PCR was performed using the LightCycler 480 system (Roche). Relative expression levels were calculated as 2-ΔCT normalized with the average CT of the housekeeping gene Tbp or Gapdh. Oligos sequences are found in supplementary table 1.
Chromatin Immunoprecipitation (ChIP)
4-6x107 mESCs were cross-linked by incubating them in DPBS (Gibco, Cat #14190094) supplemented with 1% formaldehyde (Sigma-Aldrich; Cat #252549) for 10 min rocking at room temperature. Cross-linking was stopped using 125 mM Glycine (Sigma-Aldrich, Cat #G8790) rocking for 5 minutes at room temperature. Fixed cells were then washed twice with ice-cold DPBS, and scrapped and collected using DPBS supplemented with protease/phosphatase inhibitor cocktail composed by 1X cOmplete™, EDTA-free Protease Inhibitor Cocktail (Roche, Cat #11873580001), 1 mM PMSF, 1mM Sodium Ortovanadate and 20 mM β-Glycerol phosphate. Cells were centrifuged at 3200g for 5 minutes at 4°C. Cells were lysed by resuspending cell pellet in ice-cold ChIP buffer (1 volume of SDS buffer [100mM NaCl, 50mM Tris-HCl pH 8.1, 5mM EDTA pH 8, 0.5% SDS] and 0.5 volume of triton dilution buffer [100mM Tris-HCl pH 8.6, 100mM NaCl, 5mM EDTA pH 8, 5% Triton X-100]) supplemented with protease/phosphatase inhibitor cocktail, and samples were sonicated using sonication beads (Diagenode; Cat #C01020031) and the Bioruptor Pico Sonicator (Diagenode; Cat #B01060010) for 20 cycles (each cycle 30 seconds on/30 seconds off) or till DNA fragments have 100-300bp size. Samples were centrifuged at 16000g for 20 minutes at 4°C. Chromatin was quantified, and 30 μg of chromatin was incubated with every 5μg of antibody rotating for 16 hours (or overnight) at 4°C. The following antibodies were used: H3K4me3 (Abcam, Cat #ab8580), H3K27me3 (Millipore, Cat ##07-449), H3K27Ac (Abcam, Cat #ab4729), H3K4me1 (Abcam, Cat #ab8895). Samples were pulled down by incubation with Protein A-Sepharose™ CL-4B (previously hydrated and blocked with 0.05% BSA) for 3h at 4°C in rotation. Sample was washed with a low salt washing buffer (50mM HEPES pH 7.5, 140mM NaCl and 1% Triton) three times, one wash with a high salt wash buffer (50mM HEPES pH 7.5, 500mM NaCl and 1% Triton) and one wash with TE pH 8 (10mM Tris-HCl pH8, 1mM EDTA). Samples were then eluted in 1% SDS and 100mM NaHCO3. Samples were descrosslinked by incubating for 16 hours (or overnight) at 65°C shaking at 450 rpm, and were treated with Proteinase K for 1h at 45°C. DNA was purified using QIAquick PCR Purification Kit (Qiagen, Cat #28106), following the manufacturer’s instructions.
For ChIP followed by sequencing (ChIP-seq) Purified DNA concentration and integrity were determined using Agilent Bioanalyzer (Agilent Technologies; Cat #G2939BA). Libraries were prepared using standard protocols. Chromatin was sequenced using Illumina HiSeq platform (Illumina, Inc.) (50bp single-end reads). Samples sequencing depth range was the following: (i) H3K4me3: 2.56-2.74x107 reads, (ii) H3K27me3: 6.19-7.2x107 reads, (iii) H3K27Ac: 9.11-10.65x107 reads and (iv) H3K4me1: 9.76-10.58x107 reads.
ChIP-seq data analysis
Quality control was performed on raw data with FASTQC tool. Raw reads were trimmed to remove adapters presence with Trimgalore (v0.6.6) 49. Default parameters were used except for a minimum quality of 15 (Phred score) and an adapter removal stringency of 3bp overlap. For H3K4me3, H3K27me3 and H3K4me1 data, trimmed reads were aligned to the reference genome with Bowtie2 (v2.4.4) which was executed with default parameters 50. Required genome index was built with corresponding GRCm38 fasta file retrieved from Ensembl (http://ftp.ensembl.org/pub/release-102/). Multimapped reads and those exhibiting MAPQ < 20 were removed. Randomly placed multi mappers were removed from the mapped reads. For all cases, duplicated reads were marked with SAMtools (v1.15) 51. Drosophila Melanogaster spike-ins, present in H3K27me3, were discarded. NSC and RSC quality metrics were computed with PhantomPeakQualTools (v1.2) 52. ENCODE blacklisted regions (mm10 v2) were removed prior to peak calling. BigWig files were individually generated using deepTools (v3.5.1) bamCoverage with -ignoreDuplicates -binSize 10 -smoothLength 30 - effectiveGenomeSize 2308125349 -normalizeUsing RPGC and -extendReads Fragment_Length options 53. The effective genome size was particularized to a read length of 50bp and directly retrieved from deepTools web site (https://deeptools.readthedocs.io/en/develop/content/feature/effectiveGenomeSize.html). Fragment_Length was retrieved from PhantomPeakQualTools results. For all histone marks except for H3K4me3, peak calling was conducted by means of epic2 (v0.0.52) with -effective-genome-fraction 0.8452 -fragment-size Fragment_Length options and chromosome sizes only referring to canonical chromosomes 54. For H3K4me3, MACS2 (v2.2.7.1) was used to identify peaks with -nomodel -extsize Fragment_Length -g 2308125349 options 55. The corresponding input sample was used in all peak calling computations. For the histone marks, peaks were called with adjusted p-values (FDR) < 0.05.
Peak annotation was performed with ChIPseeker R (v4.2.1) package (v1.34.1) considering a TSS region range between 5000 bp upstream and 100 bp downstream (Yu et al., 2015). Required TxDb object was generated from the Ensembl GRCm38 gtf file retrieved (http://ftp.ensembl.org/pub/release-102/). A consensus peakset was obtained from the three biological replicates per histone mark and condition (IκBα-WT or IκBα-KO) by identifying overlapping peaks in at least two out of the three replicates.
Differential binding analysis (DBA) was conducted with DiffBind (v3.8.4) per histone mark 56. Default parameters were used except for the summits parameter which was set to 500bp for all histone marks except for H3K4me3 (150bp) in order to consider, for testing, intervals of 1000bp or 300bp respectively. Summit values were selected to have interval widths between the minimum and first quartile peak width values for each histone mark. EdgeR was the statistical method used for all the three analyses. Differentially bound regions (DBRs) between IκBα-KO and IκBα-WT samples were called with adjusted p-values (FDR) < 0.05. Identified DBRs with any annotated gene were plotted with EnrichedHeatmap R package (v1.18.1) 57. To summarize replicates per condition, normalizeToMatrix function was used based on corresponding BigWig files in w0 mean mode in 50bp windows. Tracks visualization were obtained by means of Integrative Genomics Browser (IGV) tool 58.
Enhancers activity identification
Consensus peaksets derived from the three H3K27ac and H3K4me1 IκBα-WT mESCs replicates were used to identify poised and active enhancers in an IκBα-WT scenario. Putative poised enhancers were defined as those regions with H3K4me1 but no H3K27ac peaks. Putative active enhancers were defined as those regions with H3K27ac and presence/absence H3K4me1 peaks. To assess any differential enhancers activity in IκBα-KO vs IκBα-WT mESCs cultured in Serum/LIF, differentially bound regions and consensus peaksets (IκBα-KO) identified in H3K4me1 and H3K27ac were required. Putative gained or lost poised/active enhancers in IκBα -/- were obtained with following criteria: (i) for gained/lost poised enhancers in IκBα -/-: a differential H3K4me1 increase/decrease and absence of H3K27ac peaks in IκBα -/- consensus peakset, (ii) for gained/lost active enhancers (without H3K4me1) in IκBα-KO: a differential H3K27ac increase/decrease and absence of H3K4me1 in IκBα-KO consensus peakset and (iii) for gained/lost active enhancers (with H3K4me1) in IκBα-KO: a differential H3K27ac increase/decrease and presence of H3K4me1 in IκBα -/- consensus peakset.
A maximum gap of 1000bp was allowed for checking the overlap between two different histone marks. For this purpose, the ‘subsetByOverlaps’ function from the IRanges R package was used (v.2.34.1).
RNA-seq experiments
Total RNA from three independent clones from mESCs, 48h EBs and 96h EBs was isolated using the RNeasy Plus Mini Kit (Qiagen; Cat #74136) following manufacturer’s instructions. Amount of RNA was quantified with Nanodrop (Thermo Fisher; Cat #ND2000CLAPTOP), and RNA integrity was addressed by agarose gel and Agilent Bioanalyzer (Agilent Technologies; Cat #G2939BA). Libraries sequenced using Illumina HiSeq 2500 (Illumina, Inc.) (125bp paired-end reads). Samples sequencing depth ranged between 35M and 52M reads (average 41M reads) per sample.
RNA-seq data analysis
Quality control was performed on raw data with the FASTQC tool (v0.11.9). Raw reads were trimmed to remove adapter presence with Trimgalore (v0.6.6) 49. Default parameters were used except for a minimum quality of 15 (Phred score) and an adapter removal stringency of 3bp overlap. Trimmed reads were aligned to reference the genome with STAR aligner tool (v2.7.8). STAR was executed with default parameters except for the number of allowed mismatches which was set to 1. Required genome index was built with corresponding GRCm38 gtf and fasta files retrieved from Ensembl (http://ftp.ensembl.org/pub/release-102/). Obtained BAM files with uniquely mapped reads were considered for further analysis. Raw gene expression was quantified using featureCounts tool from subRead software (v2.0.1) with exon as feature 59. The raw counts matrix was imported into the R Statistical Software environment (v4.2.1) for downstream analysis. Raw expression matrix included 55,487 genes per 18 samples in total. Experimental design considered three timepoints: mouse ESCs, EBs at 48h and EBs at 96h. Each time point included 6 samples distributed in two conditions: 3 IκBα-WT and 3 IκBα-KO. Prior to statistical analysis, those genes with less than 10 raw counts across the 6 samples under test were removed. After pre-filtering, 21,843 genes (mESCs), 21,424 genes (EBs 48h) or 21,664 genes (EBs 96h) were available for testing. For visualization purposes, counts were normalized by the variance-stabilizing transformation method as implemented in DESeq2 R package 60 (v1.38.3). Differential expression analysis (DEA) was conducted with DESeq2. Each time point was independently analyzed. Fitted statistical model included sample conditions as covariable with IκBα-WT as the reference. Obtained log2 fold change values were shrunken with apeglm shrinkage estimator R package (v1.20.0) 61. Raw p-values were adjusted for multiple testing using the Benjamini-Hochberg False Discovery Rate (FDR) 62. Differentially Expressed Genes (DEGs) between IκBα-KO and IκBα-WT samples were called with adjusted p-values (FDR) < 0.05 and absolute shrunken log2 Fold change > 1. Data visualization was performed with the ggplot2 (v3.4.1).
Genome-wide DNA methylation samples preparation
DNA from frozen mESCs pellets was extracted using DNeasy Blood and Tissue Kit (Qiagen GmbH, Hilden, Germany). Purified genomic DNA was quantified with Qubit (Invitrogen, Carlsbad, CA, USA) according to manufacturer’s instructions. Infinium Mouse Methylation BeadChip (Illumina, Inc., San Diego, CA, USA) arrays were used to profile DNA methylation genome wide. This platform allows over 285,000 methylation sites per sample to be interrogated at single-nucleotide resolution. The samples were bisulfite converted using EZ DNA Methylation-Gold™ Kit (Zymo Research, CA, USA) and were hybridized in the array following the manufacturer’s instructions.
DNA methylation data analysis
The DNA methylation profile of the studied samples was assessed using the Infinium Mouse Methylation BeadChip Array (∼285,000 methylation sites) as previously described 63. Briefly, raw signal intensities were obtained with GenomeStudio Software 2011.1 (Illumina) and DNA methylation beta values were computed from raw IDAT files using GenomeStudio default normalization with control probes and background subtraction. Quality control steps to remove erratic probe signals were performed within the R statistical environment (v4.0.3). We removed probes with detection p value > 0.01, genotyping probes and manufacturing flagged (MFG) probes described in the Illumina manifest file (https://support.illumina.com/downloads/infinium-mouse-methylation-manifest-file.html). The differentially methylated probes (DMPs) between IκBα-WT and IκBα-KO samples were computed separately for each time point (mESCs and EBs at 96h) by deriving a linear model with the limma R package (v3.46.0). Each condition included 3 samples per time point. DMPs with adjusted p-value (FDR) <0.05 and absolute mean methylation beta value difference between conditions >0.3 were considered significant. DNA methylation analysis was performed using the mm10 mouse genome reference build and the complete annotation was downloaded from the annotated manifest file (http://zwdzwd.github.io/InfiniumAnnotation#mouse) 36.
Functional analysis
Overrepresentation analysis was applied over lists of selected genes derived from RNA-seq data (DEGs) or from ChIP-seq data (differentially bound regions). The Gene Ontology (Biological Process ontology, GO BP terms), KEGG PATHWAY and WikiPathways databases for Mus Musculus 64–66 were interrogated by means of clusterProfiler R package (v4.6.2) (Wu et al., 2021). Corresponding Entrez identifiers were used. Benjamini-Hochberg procedure was used to obtain adjusted p-values. Obtained GO BP terms were simplified using the simplify function from clusterProfiler with default parameters. Overrepresented terms or processes were called with adjusted p-values (FDR) < 0.05.
Gene Set Enrichment Analysis (GSEA) was performed for mESCs against the three cell states signatures defined by other authors 67. Briefly, each signature was composed of 200 genes uniquely expressed in each state obtained from bulk RNA-seq performed per state. For this purpose, the complete list of genes from mESCs samples (21,843 genes) was ranked based on the shrunken log2 Fold Change obtained from DEA. GSEA was conducted through the fgseaMultilevel function from fgsea R package (v1.24.0) 68 with default parameters. Enrichment plots were generated with the same package.
Additionally, the testEnrichment function from SeSAMe R package (v1.14.2) 69 was used for conducting functional analysis with default parameters over the list of DMPs. Probe design, transcription factor binding site and histone modifications consensus database sets, included in the same package, were interrogated.
Gene Set Variation Analysis (GSVA) R package (v1.46.0) with default parameters were used to obtain Z-score values for genes annotated to endoderm (GO:0001706), mesoderm (GO:0001707) and ectoderm (GO:0001705) GO BP terms 70.
Statistical analysis
Statistical analysis was performed with GraphPad Prism v.8.0.1. (GraphPad Software, Inc.). Unless specified, the comparison between two groups was performed with unpaired two-sided t-test. A p-value < 0.05 was considered significant.
Data availability
All sequencing data is deposited at GEO under a SuperSeries with the accession number GSE239565)[https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE239565]. Individual SubSeries can be found at: GSE239563 (RNA-seq data), GSE239564 (ChIP-seq data) and GSE239562 (Methylation array data).
Acknowledgements
We would like to acknowledge all members of the Bigas/Espinosa labs for helpful discussions. We are grateful for technical support to CRG/UPF Flow Cytometry Unit, CRG Genomics and Advanced Light Microscopy Units. This work has been supported by Spanish Ministry of Science and Innovation (PID2019-104695RB-I00, PLEC2021-007518, PDC2021-120817-I00), Generalitat de Catalunya (2021SGR 39) and Departament de Salut (SLT002/16/00299) to AB; the Spanish Ministry of Science and Innovation (PID2019-108322GB-100 and PID2022-142679NB-I00) to LDC; the Spanish Ministry of Science and Innovation (PID2021-123383NB-I00.) and the Agencia de Gestió d’Ajuts Universitaris i de Recerca (2021 SGR 01222) to BP. L.G.P has been a recipient of FI AGAUR fellowship (2019 FI-B 00151/2020 FI_B1 00130) from Generalitat de Catalunya. D.A-V was funded by the FIS fellowship (FI20/00130) from Instituto Carlos III. M.B. received funding from the Ramón Areces Foundation. M.M. is a recipient of a grant from the Instituto Carlos III, grant number CA22/00011 (co-funded by the European Social Fund Plus, ESF+ and by the European Union).
Additional information
Code availability
Scripts used to process the generated bulk RNA-seq and ChIP-seq data are available in Github repository: https://github.com/BigaSpinosaLab/PAPER_IkBa_Naive_Pluripotency
Author contribution
L.G.P, L.E and A.B designed the study. L.G.P, D.A-V, M.B, A.I, J.G, J.B, D.A.E, C.A.G.P, C.B, V.R.C and A.V performed experiments. L.G.P, M.M, M.B and C.A.G.P performed bioinformatics analysis. L.G and A.B wrote the manuscript with input from all authors. C.B, A.V, A.V, P.M, L.D.C, G.S, B.P, M.E and L.E provided input on experimental design. L.E and A.B provided resources and supervised the research.
References
- 1.The role of pluripotency gene regulatory network components in mediating transitions between pluripotent cell statesCurr. Opin. Genet. Dev 23:504–511
- 2.Heterogeneity of embryonic and adult stem cellsCell Stem Cell 3:480–483
- 3.Gene expression heterogeneities in embryonic stem cell populations: origin and functionCurr. Opin. Cell Biol 23:650–656
- 4.The exploration of pluripotency space: Charting cell state transitions in peri-implantation developmentCell Stem Cell 28:1896–1906
- 5.The Divergent Pluripotent States in Mouse and Human CellsGenes 13https://doi.org/10.3390/genes13081459
- 6.The ground state of embryonic stem cell self-renewalNature 453:519–523
- 7.The transcriptional and epigenomic foundations of ground state pluripotencyCell 149:590–604
- 8.Lineage-Specific Profiling Delineates the Emergence and Progression of Naive Pluripotency in Mammalian EmbryogenesisDev. Cell 35:366–382
- 9.Derivation of pluripotent epiblast stem cells from mammalian embryosNature 448:191–195
- 10.New cell lines from mouse epiblast share defining features with human embryonic stem cellsNature 448:196–199
- 11.Dynamic stem cell states: naive to primed pluripotency in rodents and humansNat. Rev. Mol. Cell Biol 17:155–169
- 12.Klf4 reverts developmentally programmed restriction of ground state pluripotencyDevelopment 136:1063–1069
- 13.Epigenetic differences between naïve and primed pluripotent stem cellsCell. Mol. Life Sci 75:1191–1203
- 14.Epigenetic reversion of post-implantation epiblast to pluripotent embryonic stem cellsNature 461:1292–1295
- 15.Whole-genome bisulfite sequencing of two distinct interconvertible DNA methylomes of mouse embryonic stem cellsCell Stem Cell 13:360–369
- 16.Naive pluripotency is associated with global DNA hypomethylationNat. Struct. Mol. Biol 20:311–316
- 17.Dynamic heterogeneity and DNA methylation in embryonic stem cellsMol. Cell 55:319–331
- 18.Epigenomic comparison reveals activation of “seed” enhancers during transition from naive to primed pluripotencyCell Stem Cell 14:854–863
- 19.Reorganization of enhancer patterns in transition from naive to primed pluripotencyCell Stem Cell 14:838–853
- 20.Unravelling the complexities of the NF-kappaB signalling pathway using mouse knockout and transgenic modelsOncogene 25:6781–6799
- 21.The Transcription Factor NF-κB in Stem Cells and DevelopmentCells 10https://doi.org/10.3390/cells10082042
- 22.The NF-κB family: Key players during embryonic development and HSC emergenceExp. Hematol 44:519–527
- 23.ATG5 attenuates inflammatory signaling in mouse embryonic stem cells to control differentiationDev. Cell https://doi.org/10.1016/j.devcel.2024.01.026
- 24.Nanog maintains pluripotency of mouse embryonic stem cells by inhibiting NFkappaB and cooperating with Stat3Nat. Cell Biol 10:194–201
- 25.Self-renewal versus lineage commitment of embryonic stem cells: protein kinase C signaling shifts the balanceStem Cells 29:618–628
- 26.30 Years of NF-κB: A Blossoming of Relevance to Human PathobiologyCell 168:37–57
- 27.Chromatin-bound IκBα regulates a subset of polycomb target genes in differentiation and cancerCancer Cell 24:151–166
- 28.IκBα deficiency imposes a fetal phenotype to intestinal stem cellsEMBO Rep 21
- 29.Dynamic chromatin association of IκBα is regulated by acetylation and cleavage of histone H4EMBO Rep 22
- 30.IκBα controls dormancy in hematopoietic stem cells via retinoic acid during embryonic developmentNat. Commun 15
- 31.Recruitment of IκBα to the hes1 promoter is associated with transcriptional repressionProceedings of the National Academy of Sciences 101:16537–16542
- 32.Separation-of-function mutants reveal the NF-κB-independent involvement of IκBα in the regulation of stem cell and oncogenic programsbioRxiv
- 33.The translational landscape of ground state pluripotencyNat. Commun 11
- 34.Whsc1 links pluripotency exit with mesendoderm specificationNat. Cell Biol 21:824–834
- 35.Comparative analysis of naive, primed and ground state pluripotency in mouse embryonic stem cells originating from the same genetic backgroundSci. Rep 8
- 36.DNA methylation dynamics and dysregulation delineated by high-throughput profiling in the mouseCell Genom 2https://doi.org/10.1016/j.xgen.2022.100144
- 37.Prolonged Mek1/2 suppression impairs the developmental potential of embryonic stem cellsNature 548:219–223
- 38.The RNA Helicase DDX6 Controls Cellular Plasticity by Modulating P-Body HomeostasisCell Stem Cell 25:622–638
- 39.Ancestral function of Inhibitors-of-kappaB regulates Caenorhabditis elegans developmentSci. Rep 10
- 40.Shared principles in NF-kappaB signalingCell 132:344–362
- 41.A fourth IkappaB protein within the NF-kappaB signaling moduleCell 128:369–381
- 42.IκBα controls dormancy induction in Hematopoietic stem cell development via retinoic acidbioRxiv https://doi.org/10.1101/2022.11.17.516971
- 43.Separation-of-function mutants reveal the NF-κB-independent involvement of IκBα in the regulation of stem cell and oncogenic programsbioRxiv https://doi.org/10.1101/2023.06.21.545928
- 44.The underdeveloped innate immunity in embryonic stem cells: The molecular basis and biological perspectives from early embryogenesisAm. J. Reprod. Immunol 81
- 45.Endogenous epitope-tagging of Tet1, Tet2 and Tet3 identifies TET2 as a naïve pluripotency markerLife Sci Alliance 2https://doi.org/10.26508/lsa.201900516
- 46.In vitro differentiation of mouse embryonic stem cells as a model of early hematopoietic developmentMethods Mol. Biol 538:317–334
- 47.Structural basis of seamless excision and specific targeting by piggyBac transposaseNat. Commun 11
- 48.NIH Image to ImageJ: 25 years of image analysisNat. Methods 9:671–675
- 49.TrimGalore: v0. 6.7-DOI via ZenodoZenodo https://doi.org/10.5281/zenodo.5127899.2
- 50.Fast gapped-read alignment with Bowtie 2Nat. Methods 9:357–359
- 51.Twelve years of SAMtools and BCFtoolsGigascience 10https://doi.org/10.1093/gigascience/giab008
- 52.ChIP-seq guidelines and practices of the ENCODE and modENCODE consortiaGenome Res 22:1813–1831
- 53.deepTools2: a next generation web server for deep-sequencing data analysisNucleic Acids Res 44:W160–W165
- 54.epic2 efficiently finds diffuse domains in ChIP-seq dataBioinformatics 35:4392–4393
- 55.Model-based analysis of ChIP-Seq (MACS)Genome Biol 9
- 56.DiffBind: differential binding analysis of ChIP-Seq peak dataR package version 100
- 57.EnrichedHeatmap: an R/Bioconductor package for comprehensive visualization of genomic signal associationsBMC Genomics 19
- 58.Integrative genomics viewerNat. Biotechnol 29:24–26
- 59.featureCounts: an efficient general purpose program for assigning sequence reads to genomic featuresBioinformatics 30:923–930
- 60.Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2Genome Biol 15
- 61.Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differencesBioinformatics 35:2084–2092
- 62.Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple TestingJ. R. Stat. Soc. Series B Stat. Methodol 57:289–300
- 63.Validation of a DNA methylation microarray for 285,000 CpG sites in the mouse genomeEpigenetics 17:1677–1685
- 64.Gene ontology: tool for the unification of biology. The Gene Ontology ConsortiumNat. Genet 25:25–29
- 65.KEGG: kyoto encyclopedia of genes and genomesNucleic Acids Res 28:27–30
- 66.WikiPathways: connecting communitiesNucleic Acids Res 49:D613–D621
- 67.Transcription factor antagonism regulates heterogeneity in embryonic stem cell statesMol. Cell 82:4410–4427
- 68.Fast gene set enrichment analysisbioRxiv 60012https://doi.org/10.1101/060012
- 69.SeSAMe: reducing artifactual detection of DNA methylation by Infinium BeadChips in genomic deletionsNucleic Acids Res 46
- 70.GSVA: gene set variation analysis for microarray and RNA-seq dataBMC Bioinformatics 14
Article and author information
Author information
Version history
- Preprint posted:
- Sent for peer review:
- Reviewed Preprint version 1:
Copyright
© 2025, Palma et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.