Abstract
Free full text
Xist deletional analysis reveals a co-dependency between Xist RNA and Polycomb complexes for spreading along the inactive X
SUMMARY
During X-inactivation, Xist RNA spreads along an entire chromosome to establish silencing. However, the mechanism and functional RNA elements involved in spreading remain undefined. By performing a comprehensive endogenous Xist deletion screen, we identify Repeat B as crucial for spreading Xist and maintaining Polycomb repressive complexes 1 and 2 (PRC1/PRC2) along the inactive X (Xi). Unexpectedly, spreading of these three factors is inextricably linked. Deleting Repeat B or its direct binding partner, HNRNPK, compromises recruitment of PRC1 and PRC2. In turn, ablating PRC1 or PRC2 impairs Xist spreading. Therefore, Xist and Polycomb complexes require each other to propagate along the Xi, suggesting a feedforward mechanism between RNA initiator and protein effectors. Perturbing Xist/Polycomb spreading causes failure of de novo Xi silencing, with compensatory downregulation of the active X, and also disrupts topological Xi reconfiguration. Thus, Repeat B is a multifunctional element that integrates codependent Xist/Polycomb spreading, silencing, and chromosome architecture.
ETOC BLURB
This study demonstrates that Repeat B is essential for spreading Xist, PRC1, and PRC2 along the inactive X. Spreading of Xist, PRC1, and PRC2 is inter-dependent. Without Repeat B, PRC1 and PRC2 cannot spread. Reciprocally, without PRC1 or PRC2, Xist cannot spread properly, implicating a feedforward mechanism for RNA-protein propagation.
INTRODUCTION
X-chromosome inactivation (XCI) has served as an epigenetic archetype (Starmer and Magnuson, 2009; Lee, 2011; Disteche, 2016; Mira-Bontenbal and Gribnau, 2016). During XCI, the 17-kb noncoding RNA Xist spreads exclusively in cis along the future inactive X (Xi) and induces conversion to a heterochromatic state (Brown et al., 1992; Clemson et al., 1996; Marahrens et al., 1997). The functions of Xist are manifold. On one hand, Xist acts as a modular RNA scaffold in the assembly of repressive protein factors. Two well-known factors, Polycomb repressive complexes 1 and 2 (PRC1/PRC2), are responsible for monoubiquitylating histone H2A at lysine 119 (H2AK119ub) and trimethylating histone H3 at lysine 27 (H3K27me3), respectively (Schoeftner et al., 2006; Zhao et al., 2008; Chu et al., 2015; McHugh et al., 2015; Minajigi et al., 2015; Moindrot et al., 2015; Monfort et al., 2015). On the other hand, Xist forms a repressive compartment by repelling transcriptional and architectural factors to establish a unique Xi chromosome conformation (Nora et al., 2012; Rao et al., 2014; Deng et al., 2015; Minajigi et al., 2015; Giorgetti et al., 2016).
Although broad functions have been associated with Xist, specific mechanisms have not been clarified—in particular, how Xist RNA spreads in cis. Recent work has demonstrated the importance of nuclear matrix factors in restricting Xist to the Xi territory (Hasegawa et al., 2010; Ridings-Figueroa et al., 2017; Sunwoo et al., 2017). On the Xi itself, Xist spreads from its site of transcription to nearby contacts in 3D space, preferentially targeting regions enriched for active genes before spreading to less active and gene-poor regions (Engreitz et al., 2013; Simon et al., 2013). However, the mechanism by which Xist associates with and spreads along Xi chromatin remains unknown.
Lacking in the field is a comprehensive map of Xist functional elements. While several essential domains have been identified—often corresponding to conserved repetitive motifs (“Repeats A-F”) (Wutz et al., 2002; Zhao et al., 2008; Hoki et al., 2009; Jeon and Lee, 2011; Ridings-Figueroa et al., 2017; Sunwoo et al., 2017; Yue et al., 2017)—these together account for <20% of Xist’s total sequence. Prior to the CRISPR/Cas9 technology (Ran et al., 2013), genetic dissection at the endogenous locus proved challenging due to inefficiency of homologous targeting, compounded by Xist’s large size and purported redundant regions. Previous analyses have relied heavily on the use of Xist transgenes and ectopic insertions, often in male cells (Lee et al., 1999; Wutz et al., 2002; Jeon and Lee, 2011; Pintacuda et al., 2017), with the caveat that these non-physiological perturbations might not inform Xist function in the endogenous context.
Here, we carry out a systematic deletional analysis of endogenous Xist and identify a specific RNA motif—Repeat B—for RNA spreading and Polycomb targeting. In doing so, we reveal the surprising discovery that Xist and Polycomb complexes depend on each other to spread across the Xi.
RESULTS
Comprehensive deletional analysis of native Xist in female cells
To performed a systematic CRISPR/Cas9 deletion screen, guide RNA (gRNA) pairs were designed to remove consecutive 1-2 kb regions across the Xist locus in female mouse embryonic fibroblasts (MEFs), where XCI has already been established (Fig. 1A). The transformed MEFs were tetraploid (with genome duplication after XCI) carrying two Xi’s and two Xa’s within the same nucleus (Yildirim et al., 2011), thus enabled isolation of Xi+/− clones (deletion on only one Xi) and Xi−/− clones (deletion on both Xi’s) (Fig. 1B). Xi+/− cells provided an internal control Xist cloud within the same nucleus for comparative microscopy, while Xi−/− cells provided a homogeneous system for genomic experiments. We screened for mutant clones by two-color RNA FISH—cyan probes external and red probes internal to each deletion—and selected clones exhibiting cyan with no overlapping red signal (Fig. 1B) and validated them by Sanger sequencing (File S1).
We began with a visual inspection of Xist cloud morphology by RNA FISH. Of the 13 deletions, 7 exhibited some phenotype. While Repeat A is known for its role in gene silencing (Wutz et al., 2002; Zhao et al., 2008), its deletion has been reported to cause decreased accumulation and/or loss of Xist expression in both human and mouse cells (Chow et al., 2007; Zhao et al., 2008; Hoki et al., 2009). A minimal Repeat A deletion allowed us to derive clones with an intact Xist cloud and overall RNA level (Fig. 1C, D). However, further characterization revealed aberrant splicing—as suggested previously (Royce-Tolland et al., 2010)—through desuppression of a cryptic splice donor (Fig. S1). Because this resulted in simultaneous skipping of the majority of exon 1 in ~50% of transcripts, we did not pursue the ΔRepA clones further. Likewise, exon 7a or 7d deletions caused skipping of adjacent exon 7 regions in most transcripts, exhibiting a pattern similar to Xist’s minor splice isoform (Fig. S1).
Deleting the region containing Repeat F caused loss or significant weakening of Xist clouds (Fig. 1C), consistent with other reports (Jeon and Lee, 2011; Makhlouf et al., 2014). RT-qPCR in ΔRepF Xi−/− cells confirmed a reduction in Xist RNA level (Fig. 1D), which could be due to loss of expression (Makhlouf et al., 2014), RNA stability, and/or proper “nucleation” (Jeon and Lee, 2011). Deletion of the internal exons 2-6 also yielded a weaker cloud and decreased RNA levels (Fig. 1C, D), presumably by affecting splicing efficiency and/or transcript stability.
Intriguingly, deleting Repeat B- or E-containing regions produced Xist clouds with aberrant morphologies. While ΔRepE caused widespread dispersal of Xist throughout the nucleus (Ridings-Figueroa et al., 2017; Sunwoo et al., 2017; Yue et al., 2017), ΔRepB caused Xist clouds to appear more diffuse yet remain localized near the Xi vicinity (Fig 1C). Morphological aberrations were accentuated using single-molecule super-resolution imaging by 3D stochastic optical reconstruction microscopy (3D STORM) (Fig. 1E). ΔRepB’s diffuse Xist cloud was not due to changes in Xist RNA level (Fig. 1D), nor failure to recruit the nuclear matrix protein CIZ1 as was the case for ΔRepE (Fig. S2A) (Ridings-Figueroa et al., 2017; Sunwoo et al., 2017). Thus, ΔRepB represents a distinct mechanism of Xist RNA localization. To pinpoint the specific element responsible for ΔRepB’s phenotype, we generated smaller internal deletions (Fig. S2B). The ~300 bp subdeletion of Repeat B itselt (ΔRepBd) fully recapitulated aberrant Xist clouds, whereas the other subdeletions did not. These data show that the conserved repetitive GCCCC(A/T) element itself is critical for proper Xist localization.
Next, we investigated whether any deletions might affect Xist-dependent chromatin modifications enriched on Xi. Specifically, we performed immunofluorescence (IF) for H2AK119ub and H3K27me3 marks, mediated by PRC1 and PRC2, respectively (Fig. 1F, S3). The ΔRepA Xi showed a reduction in both marks, which could be caused by loss of Repeat A and/or other exon 1 regions due to the confounding splicing defect. Similarly, ΔRepF and ΔEx2-6 showed decreased H2AK119ub and H3K27me3 Xi enrichment, presumably due to the decrease in Xist RNA levels. Strikingly, this was not the case for ΔRepB. Although Xist still partially covered the mutant Xi, there was complete loss of H2AK119ub and H3K27me3. ΔRepB contrasted sharply with ΔRepE, which caused only moderate loss of these marks despite having an even stronger impact on Xist localization (Fig. 1C, E). Therefore, Xist delocalization cannot completely account for the absence of H2AK119ub/H3K27me3 on the ΔRepB Xi. Henceforth, we focus on ΔRepB for its unique effect on both Xist localization and Xi chromatin modifications.
Genomic mapping reveals defects in Xist spreading and Polycomb recruitment
ΔRepB’s aberrant cloud morphology suggested a defect in RNA spreading. To visualize Xist binding sites on chromatin, we performed Capture Hybridization Analysis of RNA Targets (CHART-seq) in ΔRepB Xi−/− cells. Since the mutant MEFs were derived from F1 hybrid cells in which the Xi’s are of Mus musculus (mus) origin and Xa’s of Mus castaneus (cas) origin, SNPs allowed for allelic analysis of sequencing results (Yildirim et al., 2011). Antisense oligos to Xist (avoiding our deleted area) efficiently captured Xist RNA and associated Xi chromatin (Fig. 2A). Whereas WT cells displayed an expected pattern of dense chromosome-wide Xist coverage, ΔRepB Xi−/− cells showed reduced coverage across the entire Xi relative to WT. Importantly, Xist occupancy was most strongly affected at regions more distant from its site of synthesis—particularly at centromeric and telomeric ends—arguing for inefficient spreading (Fig. 2A, Δ track). Plots of Xist density indicated significantly diminished coverage over genes normally subject to XCI as well as over non-expressed genes (Fig. 2B). Examination of specific genes confirmed a reduction of Xist occupancy across gene bodies and surrounding intergenic regions (Fig. 2C). Xist coverage was reduced to the point of being indistinguishable from that over escapee genes, which normally are not subject to XCI and have low-level Xist coverage (Fig. 2B, C, Kdm5c). Similar depletion was not seen at the Xist locus (Fig. 2C, Xist), where the nascent transcript is naturally tethered to chromatin. Collectively, these data demonstrate that Repeat B is necessary for proper spreading of Xist across Xi.
We then examined ΔRepB’s effect on Xi chromatin modifications by performing allele-specific ChIP-seq in ΔRepB Xi−/− cells. Consistent with IF, there was near-complete loss of both H2AK119ub and H3K27me3 on a chromosome-wide scale, specific to the Xi (Fig. 2D). Depletion was uniform across the chromosome (Fig. 2D-F), in contrast to the non-uniform spreading defect of Xist RNA as visualized by CHART-seq. This distinction supports the idea that H2AK119ub/H3K27me3 loss is not merely a secondary effect of aberrant Xist spreading. Rather, we found it was due to failure of Xist to recruit PRC1/2, as shown by loss of EZH2 (the catalytic component of PRC2) and RYBP (a component of noncanonical PRC1) from the mutant Xi (Fig. 2G). While this work was in progress, another study using Xist transgenes reported a similar result (Pintacuda et al., 2017). Thus, Repeat B is essential for continual recruitment of Polycomb complexes during maintenance of XCI.
Xist spreading and Polycomb recruitment are mediated by HNRNPK
To identify trans factors involved in Repeat B function, we performed in vitro pulldown experiments using aptamer-tagged Repeat B RNA (Fig. 3A, S4A, B). Proteins significantly enriched over controls were identified by LC/MS (Fig. S4C, Table S1). The top candidate in all replicates was HNRNPK, a nuclear poly(C) RNA-binding protein involved in RNA processing, stability, and transport (Bomsztyk et al., 2004). Other candidates included the related HNRNPE1-3 (PCBP1-3) proteins. HNRNPK has been implicated in Xist-binding as well as interaction with PRC1 and PRC2 (Denisenko and Bomsztyk, 1997; Chu et al., 2015; Cirillo et al., 2016; Pintacuda et al., 2017). However, being a ubiquitous RNA-binding protein, it was unclear how HNRNPK might be specifically and functionally relevant to XCI. Here, we validated HNRNPK as a bona fide Xist-interacting protein in vivo. IF using two different antibodies demonstrated clear enrichment on Xi (Fig. 3B, S4D). Notably, enrichment of HNRNPK (but not HNRNPE1-3) only became visible upon pre-extraction of soluble protein prior to cell fixation (Fig. S4E, F), implying its tighter association with the Xi compartment.
Significantly, IF in ΔRepB Xi+/− cells revealed loss of HNRNPK from the mutant Xi (Fig. 3B), demonstrating Xist Repeat B is necessary for HNRNPK’s Xi-association. We then ectopically expressed Xist (full-length or exon 1) via transgene (Jeon and Lee, 2011) and found this was sufficient to stabilize HNRNPK on an autosome as well (Fig. 3C). To test whether Repeat B-HNRNPK interaction is direct, we performed electrophoretic mobility shift assay (EMSA) using recombinant protein and a synthetic RNA fragment (Fig. 3D, E). Indeed, we observed formation of multiple higher molecular weight species with increasing HNRNPK concentrations, indicative of >1 protein:RNA stoichiometry. Taken together, these data support a direct Repeat B-HNRNPK interaction in vitro and in vivo.
We sought whether HNRNPK is functionally relevant to Repeat B’s roles in Xist spreading and Polycomb recruitment in post-XCI cells. Attempts to generate stable HNRNPK knockout (KO) clones using CRISPR/Cas9 were unsuccessful—consistent with HNRNPK being essential in metazoans (Bomsztyk et al., 2004; Gallardo et al., 2015). However, we were able to temporarily ablate HNRNPK among a mixed cell population, and found deficiency of HNRNPK recapitulated ΔRepB phenotypes (Fig. S4G). To investigate the kinetics of these events, we performed siRNA knockdown (KD) of HNRNPK across a 6-day timecourse (Fig. 3F). Complete loss of H2AK119ub from the Xi was evident by day 2, although at this time, Xist clouds still appeared morphologically normal. Disrupted clouds did not fully manifest until day 4 and was not due to decreased expression of Xist or of genes known to affect Xist localization (Fig. S4H). Reduction of H3K27me3 enrichment over the Xi did not occur until day 6. No change in global H3K27me3 or H2AK119ub was observed by Western analysis at days 2, 4, or 6 (Fig. 3G), suggesting HNRNPK’s role in Polycomb maintenance is Xi-specific. Super-resolution imaging confirmed the diffuse Xist clouds in HNRNPK KD cells to be highly similar to those in ΔRepB cells (Fig. 3H, compare to to1E).1E). Collectively, these data demonstrate the importance of HNRNPK in mediating Xist spreading and Polycomb targeting across the Xi.
Co-dependency in the spreading of Xist RNA and Polycomb complexes
We wondered whether the Xist spreading defect might be linked to Polycomb loss. To test this, we generated RING1A/RING1B (the catalytic components of PRC1) double KO or EED (a core component of PRC2) KO MEFs and verified total loss of H2AK119ub or H3K27me3, respectively (Fig. 4A, File S1). In both KO cell lines, we observed surprising delocalization of Xist RNAs reminiscent of ΔRepB and HNRNPK KD/KO phenotypes, confirmed by super-resolution imaging (Fig. 4B, compare to to1E,1E, ,3H).3H). This was true in additional RING1A/B and EED KO clones (Fig. S5A, B, File S1), and was not an indirect effect of PRC1/2 depletion on Xist RNA level or on genes known to affect Xist localization (Fig. 4C). Importantly, HNRNPK’s association with Xist was unaffected, indicating that PRC1/2’s effect on cloud morphology occurs downstream of Repeat B-HNRNPK interaction (Fig. 4D).
Because Polycomb is known to play a role in chromatin compaction (Boettiger et al., 2016; Kundu et al., 2018), we asked whether diffuse Xist clouds could be a byproduct of Xi decompaction, rather than a true defect in Xist spreading. Sequential Xist RNA FISH and X-chromosome painting showed Xist clouds often exceeding the boundary of the underlying Xi territory in RING1A/B and EED KO cells—and occurring on the ΔRepB but not WT Xi in the same nucleus of ΔRepB Xi+/− cells (Fig. S5C). This observation is consistent with decreased accumulation of Xist on chromatin as seen by CHART-seq. Size measurements of WT Xi and Xa DNA territories revealed a difference of only ~20%, similar to a previous analysis in mouse cells (Giorgetti et al., 2016). This is considerably smaller than the 2-4 fold difference between WT and diffuse Xist RNA clouds in ΔRepB Xi+/−, HNRNPK KD, RING1A/B KO, and EED KO cells (Fig. 1E, ,3H,3H, ,4B).4B). Moreover, we were unable to detect any significant shift in Xi:Xa size ratio in ΔRepB Xi+/−, RING1A/B KO, and EED KO cells (Fig. S5C). These data suggest that the loss of Repeat B/Polycomb in post-XCI cells is not sufficient to reverse Xi compaction at this scale.
To investigate the Xist localization defect further, we performed RNA/DNA FISH on mitotic chromosomes from ΔRepB Xi+/−, RING1A/B KO, and EED KO cells. In this state, all chromosomes are condensed and WT Xist RNA often remained associated with the Xi in cis (Fig. S5D). However, ΔRepB Xist within the same cell was barely detectable or completely absent from its corresponding Xi. Similar Xist dissociation was seen in RING1A/B and EED KO cells. These analyses indicate that a difference in Xist’s ability to interact with chromatin, rather than Xi decompaction, likely accounts for the cloud dispersal phenotype.
To corroborate these findings at the molecular level, we performed CHART-seq using RING1A/B and EED KO cells. In agreement with RNA FISH analysis, Xist binding was reduced across the Xi relative to WT in both cell lines (Fig. 4E). Interestingly, EED KO showed a stronger effect than RING1A/B KO, indicating that spreading of Xist RNA may depend on PRC2/H3K27me3 more than on PRC1/H2AK119ub. Overall coverage profiles mirrored that of ΔRepB Xi−/− CHART-seq (compare to Fig. 2A), exhibiting particularly diminished Xist binding at chromosome extremities. Plots of Xist density again showed low coverage over genes normally subject to XCI and non-expressed genes (Fig. 4F). Examination of specific genes confirmed a reduction of Xist occupancy across gene bodies and surrounding intergenic regions, but not at the Xist locus itself or escapees (Fig. 4G). Depletion of PRC1 or PRC2 therefore phenocopies deletion of Repeat B. Taken together, our data reveal the surprising conclusion that, while Xist RNA recruits Polycomb complexes to the Xi, PRC1 and PRC2 are in turn required to properly spread Xist.
Independent and interdependent recruitment of Polycomb complexes
An ongoing debate in the XCI field regards the order in which PRC1 and PRC2 are recruited to chromatin (Schoeftner et al., 2006; Zhao et al., 2008; Margueron and Reinberg, 2011; Simon and Kingston, 2013; Cooper et al., 2016; Almeida et al., 2017; Pintacuda et al., 2017). Given our KO cell lines, we examined reciprocal effects of depleting PRC1 and PRC2. As shown by IF, KO of EED expectedly caused total loss of H3K27me3, but also reduced H2AK119ub enrichment on the Xi (Fig. 5A, S5B). This effect is consistent with the canonical Polycomb pathway whereby H3K27me3 facilitates PRC1 recruitment (Cao et al., 2002; Margueron and Reinberg, 2011; Simon and Kingston, 2013). Similarly, RING1A/B KO caused total loss of H2AK119ub, but also significantly reduced H3K27me3 enrichment on the Xi. This effect is consistent with the non-canonical Polycomb recruitment pathway whereby H2AK119ub facilitates PRC2 recruitment (Tavares et al., 2012; Kalb et al., 2014; Cooper et al., 2016). These reciprocal effects are consistent with the 3-way inter-dependency between Xist, PRC1, and PRC2 for continual recruitment and propagation along the Xi. Thus, the inter-dependency for spreading of Polycomb complexes may, in fact, partially reflect the requirement of an RNA. In turn, Xist delocalization may contribute toward the overall reduction of H2AK119ub/H3K27me3 in each KO cell.
Nevertheless, in both EED and RING1A/B KO cell lines, a notable fraction of cells retained H2AK119ub or H3K27me3 Xi enrichment, respectively (Fig. 5A, S5B), suggesting PRC1 and PRC2 can be recruited independently of one another to a degree. This was unexpected given a recent model in which PRC2 recruitment to the Xi was proposed to strictly depend on prior H2AK119ub modification by non-canonical PRC1 (Cooper et al., 2016; Almeida et al., 2017; Pintacuda et al., 2017). We ruled out the possibility that residual H2AK119ub/H3K27me3 Xi enrichment could be due to channel bleed-through from concurrent Xist RNA FISH, as IF alone showed identical results (Fig. S5E). To see whether the more compact state of the Xi could cause H2AK119ub or H3K27me3 to appear enriched above neighboring chromatin, we performed IF using panH2A or panH3 antibodies, respectively, in WT cells and saw no such enrichment (Fig. S5F). Taken together, perturbing either PRC1 or PRC2 weakened the other’s modification of Xi chromatin, though PRC1’s effect on H3K27me3 was stronger than PRC2’s on H2AK119ub. Yet at the same time, the two can function independently on Xi to an extent, since many cells retained weak H2AK119ub or H3K27me3 enrichment when PRC2 or PRC1 was depleted, respectively.
To confirm these results with another approach, we performed ChIP-seq for H3K27me3 in RING1A/B KO and H2AK119ub in EED KO cells. In agreement with our IF observations, EED KO caused significant reduction of H2AK119ub on Xi relative to WT, and RING1A/B KO caused even stronger reduction of H3K27me3 (Fig. 5B). These effects were chromosome-wide. Nevertheless, H2AK119ub/H3K27me3 levels remained slightly higher on Xi over Xa in PRC2/1 KO cells, respectively, as well as over Xi in ΔRepB Xi−/− cells, which showed near-complete loss of both marks (Fig. 5B-D, compare to Fig. 2). This residual enrichment was particularly noticeable over genes subject to XCI (Fig. 5C, D, Pak3) but less so over non-expressed genes (Fig. 5C, D, Dcx), which often already contain H2AK119ub/H3K27me3 on Xa. Together, these data support both independent and interdependent mechanisms of PRC1/2 recruitment during maintenance of XCI.
Failure of Xi gene silencing with partial compensatory downregulation of Xa
To determine the role of Repeat B in maintenance of gene silencing, we performed RNA-seq in ΔRepB Xi−/− MEFs. No major changes occurred in X-linked gene expression (Fig. S6A, B), despite chromosome-wide depletion of H2AK119ub and H3K27me3 (Fig. 2D). These findings are consistent with gene silencing being generally stable in post-XCI cells, independent of Xist RNA (Brown and Willard, 1994; Yildirim et al., 2013). To determine if Repeat B is required for de novo silencing, we generated the same deletion in female mouse embryonic stem (ES) cells (Fig. S6C, File S1), which undergo XCI as they differentiate. Importantly, our ES cell line is a mus/cas hybrid that selectively inactivates the mus X chromosome (Fig. S6D) (Ogawa et al., 2008), facilitating downstream allelic analysis. When differentiated for 14 days, the ΔRepB ES cells recapitulated the defective Xist cloud and H2AK119ub/H3K27me3 phenotypes seen in MEFs (Fig. 6A). Thus, Repeat B is also required for Xist spreading and Polycomb targeting during de novo establishment of XCI.
Transcriptomic analysis at day 14 of differentiation showed an upregulation of X-linked genes in ΔRepB cells compared to WT, whereas no significant change was observed for autosomes such as chromosome 13 (Fig. 6B, C, “comp”). Examination of individual alleles showed a drastic increase specific to Xi, as evidenced by a rightward shift in cumulative distribution frequency (CDF) plots and an upward deviation in scatterplots (Fig. 6B, C, “mus”). The fraction of reads from Xi shifted from ~5% (monoallelic) in WT to ~35% (biallelic) in ΔRepB ES cells (Fig. 6D). This percentage remained at the expected 50% (biallelic) for reads from chromosome 13 in both WT and ΔRepB cells. Genes that failed to be silenced showed no obvious clustering along the chromosome (Fig. 6E). Inspection of individual genes confirmed the presence of reads from the Xi in ΔRepB but not WT cells (Fig. 6F). Interestingly, Xi upregulation was accompanied by a slight but significant (p=5.7e-15) and reproducible downregulation of Xa (Fig. 6B, C, “cas”), suggesting partial Xa compensation for failed Xi silencing. This offers one possible explanation for the continued survival of ΔRepB cells despite undergoing incomplete XCI. Similar results were obtained using a second independently derived ΔRepB clone (Fig. S6E-I). Thus, Repeat B is essential during differentiation for establishing chromosome-wide gene silencing on the Xi—which if compromised, may be partially rescued by downregulating the Xa.
Deleting Repeat B impairs Xi topological reconfiguration
XCI is accompanied by architectural reconfiguration of the Xi, characterized by weakening of topologically associating domains (TADs) and formation of megadomains (Nora et al., 2012; Rao et al., 2014; Deng et al., 2015; Minajigi et al., 2015; Giorgetti et al., 2016). We wondered how deleting Repeat B might broadly affect Xi chromosome structure. To address this, we performed allele-specific in situ Hi-C in ΔRepB ES cells after 14 days of differentiation. Contact heatmaps at 100-kb resolution showed that, as expected, the WT Xi folded into two megadomains (Fig. 7A, upper left panel, “WT”). Interestingly, megadomains were also present on ΔRepB Xi (Fig. 7A, upper left panel, “ΔRepB”), despite the significant defect in XCI establishment (Fig. 6, S6E-I). Thus, while megadomain architecture is not required for Xi gene silencing (Darrow et al., 2016; Giorgetti et al., 2016; Froberg et al., 2018), our data show it is neither sufficient for, nor a consequence of gene silencing. By examining the Pearson correlation map for interaction frequencies, the two megadomains appeared even more pronounced on the Xi in ΔRepB relative to WT (Fig. 7A, lower left panel, compare “WT” and “ΔRepB”). Consistent with this observation, interaction frequency between the two megadomains on Xi decreased from 15% of total chromosome interactions (WT) to 9% (ΔRepB), whereas the Xa remained unchanged (9%). ΔRepB Xi−/− MEFs showed the same trend (Fig. S7A)—17% (WT) to 12% (ΔRepB) on Xi; Xa unchanged (12%)—suggesting Repeat B might play a role in promoting long-range chromosomal interactions spanning the megadomain border.
Next, we examined Xi TAD organization. Consistent with a recent study from our lab (Wang et al., 2018), TADs were still present on Xi but in a significantly attenuated state relative to Xa (Fig. 7B, C). However, TADs were visibly less attenuated on ΔRepB Xi compared to WT. To quantify this, we calculated “insulation scores” at 100-kb resolution across WT Xa and used these to call TADs. Consistent with prior studies (Giorgetti et al., 2016), ~110 TADs were identified (~90 after removal of regions with low SNP density). We then assigned a metric to each, with a greater “TAD score” signifying greater TAD strength (see “Methods” for details). Averaged across the chromosome, TADs were stronger on the ΔRepB Xi compared to WT (Fig. 7B), with ~30 individual TADs being significantly stronger (~8 were weaker) (Fig. 7C). We did not detect any significant correlation between strength of these TADs and corresponding gene expression (Fig. S7D), in line with our earlier observation that genes no longer silenced in ΔRepB ES cells do not cluster along the chromosome (Fig. 6E, S6H). Allele-specific in situ Hi-C in ΔRepB Xi−/− MEFs revealed similar, albeit subtler, impacts of deleting Repeat B on maintenance of Xi chromosome structure (Fig. S7A-C), despite no significant change in gene expression (Fig. S6A, B). In particular, ~20 TADs were stronger on the ΔRepB Xi compared to WT (Fig. S7B, C), similar to our previous observation in MEFs harboring a full-length Xist deletion (Minajigi et al., 2015). Collectively, these results suggest that Repeat B is at least partly responsible for Xist’s role in disrupting TADs during both establishment and maintenance of XCI. Furthermore, they demonstrate that TADs and megadomains can co-exist on the X chromosome.
In addition to TADs, mammalian chromosomes are partitioned into alternating A/B compartments, formed through self-association of active gene-rich (A) and inactive gene-poor (B) regions (Lieberman-Aiden et al., 2009; Bickmore and van Steensel, 2013; Rao et al., 2014; Bonev and Cavalli, 2016). During formation of the Xi, A/B compartments are partially merged to create larger “S1/S2” transitional compartments, which are merged further to yield the final Xi-specific macro-structure (Wang et al., 2018). To examine potential effects of deleting Repeat B on compartmentalization, we again examined Pearson correlation maps of our Hi-C interaction matrices. As expected, the Xa displayed a checkerboard pattern characteristic of A/B compartmentalization (Fig. 7A, lower right panel; S7E). Meanwhile, the WT Xi displayed a partial checkerboard pattern resembling S1/S2 compartmentalization (Fig. 7A, lower left panel, “WT”; S7E). However, our principal component analysis only detected megadomains, perhaps due to the late differentiation timepoint of our dataset (S1/S2 compartmentalization was originally reported using day 7 ES cells). This was absent on ΔRepB Xi, which instead retained Xa-like features in some regions (Fig. 7A, arrows). Taken together, our data show that Repeat B plays a critical role in transforming the X from an active to inactive chromosomal structure.
DISCUSSION
Here, we have performed the first comprehensive deletional analysis of Xist RNA in the native context and found that Repeat B plays a critical role in propagating Xist RNA along the Xi. Unexpectedly, our analyses revealed a profound inter-dependency between Xist RNA and Polycomb complexes during the process of spreading. These findings shift our view of the relationship between Xist and Polycomb from one of unidirectional to bidirectional recruitment. Indeed, our study shows that the dynamics of Polycomb recruitment are more complex than previously suggested. Both canonical (PRC2 action recruits PRC1) and non-canonical (PRC1 action recruits PRC2) pathways have been proposed, with prevalent models positing that one complex depends on the other in a hierarchical fashion. However, more recent evidence suggests a greater degree of interdependency between PRC1 and PRC2 (Schoeftner et al., 2006; Margueron and Reinberg, 2011; Kahn et al., 2016; Almeida et al., 2017; Dorafshan et al., 2017). Consistently, our results for the X chromosome show that depleting PRC2 limits but does not abolish PRC1 recruitment, and vice versa. Thus, PRC1 and PRC2 reinforce each other’s occupancy but can be recruited independently of the other to a limited extent. Our model therefore proposes that the two pathways can work in parallel during maintenance of XCI, though both require Xist Repeat B and HNRNPK.
In the simplest model (Fig. 7D,E), Xist recruits PRC1 and PRC2 to Xi chromatin. PRC2 then trimethylates H3K27 and PRC1 ubiquitylates H2AK119. The H3K27me3 and H2AK119ub marks would then serve as binding sites for PRC1 and PRC2 through the canonical and non-canonical pathways, respectively (Margueron and Reinberg, 2011; Tavares et al., 2012; Simon and Kingston, 2013; Kalb et al., 2014; Cooper et al., 2016). In this way, PRC1 and PRC2 reinforce each other’s recruitment by Xist RNA and partially self-propagate along the chromatin in cis. In turn, as revealed in this study, PRC1 and PRC2 reciprocally enable Xist to spread on the Xi in a feed-forward loop. Alternatively, Polycomb-mediated changes to chromatin structure (Francis et al., 2004; Kundu et al., 2018; Oksuz et al., 2018) may allow Xist to better interact with chromatin or spread to less accessible regions (Fig. 7D). Thus, the inter-dependency between Xist and Polycomb complexes would underlie the chromosome-wide propagation of the silencing machinery.
Notably, previous studies with PRC1/2 KOs or deletions encompassing Repeat B did not exhibit noticeable effects on the morphological character of Xist clouds (Wutz et al., 2002; Schoeftner et al., 2006; da Rocha et al., 2014; Almeida et al., 2017; Pintacuda et al., 2017). A number of reasons could explain this discrepancy, including non-native conditions employed by previous studies, such as the use of transgenes and/or forced Xist expression. Prior studies also did not employ super-resolution imaging. Here, all experiments were performed in the native context and in female cells, and furthermore benefited from inclusion of an unperturbed Xist cloud in the same nucleus for side-by-side comparison.
Another interesting result from our screen was that many of Xist’s functional elements correspond to repetitive motifs, which may act as multicopy binding platforms for trans factors. As a proof of principle, we demonstrate that Repeat B can indeed accommodate multiple molecules of HNRNPK. Previous studies have suggested a direct interaction between HNRNPK and PRC1 (Pintacuda et al., 2017) as well as PRC2 (Denisenko and Bomsztyk, 1997). Because HNRNPK is a general RNA-binding protein, HNRNPK function is likely to require cooperation with Xi-specific factors to carry out its role in Polycomb recruitment. Repeat B may be only one such Xi-specific motif. Deleting Repeat B causes substantial failure of de novo gene silencing chromosome-wide. However, partial silencing still occurred, likely due to intact Repeat A, a motif known to be involved in gene silencing (Wutz et al., 2002). Thus, Repeats A and B may work together to establish, spread, and maintain epigenetic silencing. Unfortunately, we were unable to investigate Repeat A’s individual contribution in this study due to the confounding effect on Xist splicing.
Finally, our study also demonstrates that Repeat B plays an important role in architectural reorganization of the Xi. Deleting Repeat B post-XCI resulted in failed attenuation of TADs, but at the same time left megadomains strongly intact. This effect was even greater when deleting Repeat B during de novo XCI, consistent with the accompanying failure of gene silencing. These results indicate that TADs and megadomains are not mutually exclusive, and furthermore that the Xi-specific megadomain structure does not preclude gene expression. Moreover, deleting Repeat B led to a relative decrease in interaction frequency between the two megadomains. Thus, Repeat B (and Xist in general) may play a role in promoting long-range chromosomal interactions that span the two domains. Lastly, compartment structures were also affected, with the mutant Xi lacking patterns resembling S1/S2 compartmentalization, unlike the WT Xi. Instead, the mutant Xi exhibited regional Xa-like features. Thus, Repeat B may play a role in S1/S2 compartment formation. Our study has thus identified a multifunctional RNA domain that coordinates co-dependent spreading of Xist and Polycomb complexes, gene silencing, and the topological transformation from active to inactive X chromosome structure.
STAR METHODS
Contact for Reagent and Resource Sharing
Further information and requests for reagents should be directed to and will be fulfilled by the Lead Contact, Jeannie T. Lee (ude.dravrah.hgm.oiblom@eel).
Experimental Model and Subject Details
Cell lines
The M. musculus/M. castaneus F1 hybrid transformed MEF cell line was previously described as “EY.T4” (Yildirim et al., 2011). The male and female transgenic Xist MEF cells lines carrying either full length Xist or Xist exon 1 were previously described as “♂X+P” and “♀X+P E1”, respectively (Jeon and Lee, 2011). MEFs were grown in medium containing DMEM, high glucose, GlutaMAX supplement, pyruvate (Thermo Fisher Scientific), 10% FBS (Sigma), 25 mM HEPES pH 7.2-7.5 (Thermo Fisher Scientific), 1x MEM non-essential amino acids (Thermo Fisher Scientific), 1x Pen/Strep (Thermo Fisher Scientific), and 0.1 mM βME (Thermo Fisher Scientific) at 37°C with 5% CO 2. The M. musculus/M. castaneus F2 hybrid ES cell line carrying a mutated Tsix allele was previously described as “TsixTST/+” (Ogawa et al., 2008). ES cells were grown on γ-irradiated MEF feeders in medium containing DMEM, high glucose, GlutaMAX supplement, pyruvate, 15% Hyclone FBS (Sigma), 25 mM HEPES pH 7.2-7.5, 1x MEM non-essential amino acids, 1x Pen/Strep, 0.1 mM βME, and 500 U/mL ESGRO recombinant mouse Leukemia Inhibitory Factor (LIF) protein (Sigma, ESG1107) at 37°C with 5% CO 2. LIF was excluded from the medium during ES cell differentiation procedures (see Method Details below).
Method Details
ES cell differentiation
Undifferentiated ES cells were grown on γ-irradiated MEF feeders for 3 days, after which ES cell colonies were trypsinized and feeders removed (day 0). ES cells were then switched to medium lacking LIF and grown in suspension for 4 days, forming embryoid bodies (EBs). EBs were settled down onto gelatin-coated coverslips at day 4 and allowed to further differentiate until harvesting at day 14.
Generation of Xist deletions and HNRNPK, RING1A/B, and EED KO cells
Xist deletions were generated by CRISPR-Cas9 using a pair of gRNAs flanking each target region. HNRNPK, RING1A/B, and EED KOs were also generated by CRISPR-Cas9 but using a single gRNA to disrupt the coding sequence (premature stop or frameshift mutation). gRNA sequences (Table S2) were designed using tools available online (http://crispr.mit.edu) and cloned into pSpCas9(BB)-2A-GFP or pSpCas9(BB)-2A-Puro vectors (Ran et al., 2013). gRNA-Cas9 plasmid was delivered into ES cells by electroporation (Bio-Rad Gene Pulser Xcell) or MEFs by nucleofection (Lonza Nucleofector II, using MEF 2 solution) as per manufacturer’s instructions. Following plasmid delivery, cells were cultured for one week to allow sufficient time for DNA cutting and repair. Single cells were then sorted into 96-well plates by FACS, expanded, and screened by genomic PCR, Sanger sequencing (File S1), and Xist RNA FISH (Xist deletions) or HNRNPK/H2AK119ub/H3K27me3 IF (HNRNPK, RING1A/B, EED KOs). Multiple clones were examined for each deletion/KO to rule out any effect arising from clonal variation.
Labeling of Xist oligo FISH probes
Xist oligo FISH probe sequences (Table S3) were designed using tools available online (https://www.idtdna.com/calc/analyzer). Amino-ddUTP (Kerafast) was added to the 3’-ends of pooled oligos by Terminal Transferase (New England BioLabs) as per manufacturer’s instructions. Oligos were purified by phenol/chloroform extraction, concentrated by ethanol precipitation, resuspended in 0.1 M sodium borate, and labeled with Atto488 (Sigma), Cy3B (GE Healthcare), or Alexa647 NHS-ester (Life Technologies). After another ethanol precipitation, labeled oligos were resuspended in water and labeling efficiency was evaluated by absorbance using NanoDrop (Thermo Fisher Scientific).
RNA FISH
RNA FISH was performed as previously described (Sunwoo et al., 2015). Briefly, cells were grown on glass coverslips and rinsed in PBS. They were fixed in 4% paraformaldehyde for 10 min and then permeabilized in PBS/0.5% Triton X-100 for another 10 min at room temp. Cells were rinsed in PBS and dehydrated in a series of increasing ethanol concentrations. Labeled oligo probe pool (0.1-5 nM each) was added to hybridization buffer containing 25% formamide, 2x SSC, 10% dextran sulfate, and nonspecific competitor (0.1 mg/mL mouse Cot-1 DNA [Thermo Fisher Scientific] or 1 mg/mL yeast tRNA [Thermo Fisher Scientific]). Hybridization was performed in a humidified chamber at 37°C for >3 ho urs or overnight. After being washed once in 25% formamide/2x SSC at 37°C for 20 min and thre e times in 2x SSC at 37°C for 5 min each, cells were mounted for wide-field fluorescent imaging. Nuclei were counter-stained with Hoechst 33342 (Life Technologies).
RNA FISH/X-chromosome painting
1×105 cells were cytospun onto glass slides and RNA FISH performed as described above. Slides were mounted and images captured with positions recorded. After imaging RNA signal, coverslips were carefully removed and slides rinsed first in PBS/0.05% Tween-20 and then in PBS to remove mounting medium, treated with RNase A (0.5 mg/mL in PBS) at 37°C for 40 min to remove RNA signal, and denatured for DNA FISH in 70% formamide/2x SSC at 80°C for 15 min. Slides were quenched in ice cold 70% ethanol and dehydrated in a series of increasing ethanol concentrations. 1:10 (v/v) XMP X Green mouse chromosome paint (MetaSystems, D-1420-050-FI) was added to hybridization buffer containing 50% formamide, 2x SSC, 10% dextran sulfate, and 0.2 mg/mL mouse Cot-1 DNA (Thermo Fisher Scientific) and denatured at 95°C for 10 min. Hybridization was performed in a h umidified chamber at 37°C overnight. After being washed once in 0.2x SSC at 65°C for 10 min an d three times in 2x SSC at room temp for 5 min each, slides were remounted and reimaged at recorded positions.
IF/RNA FISH
IF was performed as previously described (Sunwoo et al., 2015). Briefly, cells were grown on glass coverslips and rinsed in PBS. They were fixed in 4% paraformaldehyde for 10 min and then permeabilized in PBS/0.5% Triton X-100 for another 10 min at room temp. To detect association of HNRNPK with Xi, cells were pre-extracted with CSKT buffer (100 mM NaCl, 300 mM sucrose, 10 mM PIPES, 3 mM MgCl2, 0.5% Triton X-100, pH 6.8) for 10 min prior to fixation (no subsequent permeabilization step necessary). After being blocked for 30 min in PBS/1% BSA supplemented with 10 mM ribonucleoside vanadyl complex (New England BioLabs), primary antibodies were added and allowed to incubate at room temp for 1 hr. Cells were washed three times in PBS/0.05% Tween-20 at room temp for 5 min each. After incubating with dye-conjugated secondary antibody for 30 min at room temp, cells were washed again in PBS/0.05% Tween-20 at room temp for 5 min each. Cells were post-fixed in 4% paraformaldehyde and dehydrated in a series of increasing ethanol concentrations. RNA FISH was then performed as described above.
Preparation of mitotic chromosomes
MEFs were grown to ~75% confluency. KaryoMAX colcemid solution (Thermo Fisher Scientific) was added to cells at a final concentration of 100 ng/mL for 2 hours. Afterwards, the plate was tapped several times against a hard surface and rinsed with PBS to dislodge mitotic cells. The medium/PBS was collected and cells pelleted by centrifugation at 400 g for 5 min. The supernatant was aspirated and cells gently resuspended in 200 μL PBS. Hypotonic solution (75 mM KCl) was slowly added to a final volume of 10 mL while swirling, and cells were incubated at 37°C for 10 min. Cells were pelleted by centrifugation at 400 g for 5 min. The supernatant was aspirated and cells gently resuspended in 200 μL hypotonic solution. Cold fixative (3:1 methanol:acetic acid) was slowly added to a final volume of 10 mL while swirling, and cells were fixed at −20°C for 1 hour. Cells were pelleted by centrifugation at 400 g for 5 min, washed with an additional 10 mL of cold fixative, pelleted again, and gently resuspended in 1 mL fixative. 100 μL of fixed cell solution was dropped (dropwise) onto slides from 1 inch height, allowing the slide to air dry for 30 seconds between each drop. Slides were allowed to air dry completely for 1 hour before proceeding to RNA FISH.
Microscopy
For wide-field fluorescent imaging, cells were observed on a Nikon 90i microscope equipped with 60x/1.4 N.A. VC objective lens, Orca ER CCD camera (Hamamatsu), and Volocity software (Perkin Elmer).
3D STORM super-resolution microscopy
STORM imaging was performed as previously described (Sunwoo et al., 2015) on an N-STORM (Nikon) equipped with 100x/1.4 N.A. λ objective lens, ion X3 EM CCD camera (Andor), and 3 laser lines (647 nm, 561 nm, and 405 nm). A cylindrical lens was inserted into the optical path to introduce astigmatism. Z calibration was performed using 100 nm TetraSpeck beads (Life Technologies). Imaging buffer containing 147 mM βME and GluOX (Sigma) was used to promote blinking and reduce photo-bleaching. Z calibration was performed using 100 nm TetraSpeck beads. N-STORM module in Element software (Nikon) was used to control microscopes, acquire images, and perform 3D STORM localizations.
Microscopy image analysis
For measuring the size of Xist clouds or X chromosome territories, FISH images captured on an epifluorescent microscope were imported into ImageJ (NIH, v1.52a) using the Bio-Formats Importer plug-in. After maximum intensity projection along the z-axis, Xist clouds or X chromosome territories were outlined by the free hand selection tool and area within measured.
For alignment of Xist RNA FISH and X-chromosome painting images, the two images were cropped and saved as TIF files. TIF Images were imported into MatLab (MathWorks). Nuclei (stained with Hoechst 33342) were aligned by applying three functions: imregconfig (configuring intensity-based registration), imregtform (estimating geometric transformation for aligning images), and imwarp (applying geometric transformation). Registered image was saved as TIF.
For comparing relative intensity between different RNA FISH probes in ΔRepA MEFs, RNA FISH was performed as described above using three differently colored probe sets: Xist exon 7 (Atto488), Repeat A (Cy3B), and Repeat F (Alexa647). All measurements were done using Volocity software (Perkin Elmer). Xist clouds were identified based on exon 7 signal, and intensities for all three probe sets were recorded. Values were imported to Excel and baseline signal was subtracted based on nuclear background FISH intensity per volume. Adjusted intensity per channel for each Xist cloud was normalized to that of exon 7 in order to approximate relative level between clouds.
Antibodies
The following primary antibodies were used for ChIP-seq: H3K27me3 (GeneTex, GTX60892), H2AK119ub (Cell Signaling, CST8240); for IF: H3K27me3 (GeneTex, GTX60892), H3K27me3 (Active Motif, AM39535), H2AK119ub (Cell Signaling, CST8240), EZH2 (Cell Signaling, CST3147), HNRNPK (Proteintech, 11426-1-AP), HNRNPK (Bethyl Laboratories, A300-674A), HNRNPE1 (Proteintech, 14523-1-AP), HNRNPE2 (GeneTex, GTX114616), HNRNPE3 (GeneTex, GTX118656), CIZ1 (in-house (Sunwoo et al., 2017)); for Western blot: H3K27me3 (GeneTex, GTX60892), H3K27me3 (Cell Signaling, CST9733), H2AK119ub (Cell Signaling, CST8240), GAPDH (Cell Signaling, CST14C10), HNRNPK (Proteintech, 11426-1-AP). Dye-conjugated secondary antibodies were purchased from Life Technologies.
Western blot
Cells were washed once with PBS and lysed in cold lysis buffer (50 mM Tris pH 7.5, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS, 1x protease inhibitor cocktail [Sigma, P8340]). Lysate was sonicated (Qsonica Q800 Sonicator) in polystyrene tubes at 45% power setting, 30 sec on/30 sec off for a total sonication time of 5 min at 4°C. After removing debris by centrifugation at 16,000 g for 10 min, protein concentration in the supernatant was measured (Pierce BCA Assay Kit). 20-50 μg protein lysate was denatured in 1x Laemmli buffer at 95°C for 10 min and resolved by SDS-PAGE. Protein was electrotransferred to Immobilon-P PVDF membrane (EMD Millipore). At room temp, the membrane was blocked with blocking buffer (PBS/0.05% Tween-20 containing 5% milk) for 1 hr, incubated with primary antibody in blocking buffer for 1 hr, washed three times with PBS/0.05% Tween-20 for 5 min each, incubated with secondary antibody-HRP conjugate (Promega, W4011 or W4021) in blocking buffer for 30 min, and washed three times again with PBS/0.05% Tween-20 for 5 min each. Protein bands were visualized using Western Lightning Plus-ECL (PerkinElmer) and exposing to BioMax MR film (Carestream Health).
Southern blot
ES cells were lysed in lysis buffer (10 mM Tris pH 7.5, 10 mM NaCl, 10 mM EDTA, 0.5% sarkosyl) supplemented with 1 mg/ml proteinase K (Sigma) overnight at 55°C. Genomic DNA was precipitated by adding 2x volume of 66 mM NaCl/ethanol and washed with 70% ethanol. Restriction digestion with NcoI (New England BioLabs) was performed overnight at 37°C. 10 μg of digested DNA was resolved by agarose gel electrophoresis. DNA was de-purinated using 0.2 N HCl for 10 min, washed in deionized water, neutralized with 0.4 N NaOH, and transferred to Hybond-XL membrane (GE Healthcare) by capillary action in 0.4 N NaOH overnight at room temp. Membrane was rinsed briefly with 2x SSC and then pre-hybridized in Church buffer (7% SDS, 158 mM NaH2PO4, 342 mM Na2HPO4, 1% BSA, 1 mM EDTA) for 2 hours at 65°C. The probe was a ~500-bp PCR fragment (primers: CCTAAATGTCCTATAATCCATTGCTACACA and CGCTTGGTGGATGGAAATATGGTTTTG) labeled using Random Primers DNA Labeling System (Thermo Fisher Scientific) as per manufacturer’s instructions. Labeled probe was denatured at 95°C for 10 min prior to hybridization with membrane at ~1×106 cpm/mL overnight at 65°C. Membrane was rinsed briefly with 2x SSC, then washed twice with 2x SSC/0.1% SDS at 65°C for 20 min each, and again with 0.1x SSC/0.1% SDS at 65°C for 10 min. Membrane was exposed to BioMax MR film (Carestream Health) at −80°C for two days prior to developing.
In vitro RNA pulldown and mass spectrometry
The in vitro RNA pulldown was performed as previously described (Leppek and Stoecklin, 2014), with additional modifications. Four copies of the S1m aptamer each separated by a short poly(A) spacer were cloned downstream of a T7 promoter into pUC19 vector (New England BioLabs). The S1m aptamer is a variation of the original S1 aptamer (Srisawat and Engelke, 2001), in which the stem structure was lengthened by 8 bp, and 3 nt altered to strengthen stem formation. We further modified these 8 bps so they differ between each of the 4 tandem aptamers, thereby promoting intra- over intermolecular aptamer folding and improving pulldown efficiency. Our bait sequence (sense RepB, antisense RepB, minimal RepBd, or no insert) was cloned downstream of the tandem aptamer tag, vector linearized by restriction digest, in vitro transcribed by T7 RNA polymerase (MEGAscript, Thermo Fisher Scientific), and purified using TRIzol Reagent (Thermo Fisher Scientific). The size and quality of the RNA was verified by denaturing RNA gel electrophoresis. For each pulldown, 50 pmol of RNA were added to 200 μL binding/lysis buffer (15 mM βME, 2 mM MgCl2, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS, 1x PBS) supplemented with 200 U/mL RNase Inhibitor (Roche), and folded by incubation for 5 min each at 50°C, 37°C, then room temp. Folde d RNA was added to 20 μL High Performance Streptavidin Sepharose Beads (GE Healthcare) pre-washed with binding/lysis buffer, and allowed to bind for 2 hr with rotation at 4°C. Unbound RNA was removed by washing beads twice with binding/lysis buffer.
Protein lysate was prepared by two different methods with similar results: (1) For each pulldown experiment, 3 confluent 15-cm dishes of MEFs (~2×107 cells) were rinsed with PBS, extracted with cold CSKT buffer (10 mM PIPES pH 6.8, 100 mM NaCl, 300 mM sucrose, 3 mM MgCl2, 0.5% Triton X-100) for 10 min to remove much of the soluble protein fraction, and rinsed again with PBS. Cells were scraped, pelleted, and snap frozen until later use. (2) Cells were trypsinized, pelleted, rinsed with PBS, and resuspended in 10 mL nuclear isolation buffer (10 mM HEPES pH 7.5, 10 mM KCl, 0.1 mM EDTA, 1 mM DTT, 0.5% NP-40) supplemented with 1x complete mini EDTA-free protease inhibitor cocktail (Roche), for 20 min on ice. Nuclei were pelleted at 1,000 g for 5 min, rinsed three times with nuclear isolation buffer to remove remaining cytoplasmic debris, and snap frozen until later use. To prepare lysate, extracted cells/nuclei were thawed on ice, resuspended in 200 μL cold binding/lysis buffer supplemented with 2 μL protease inhibitor cocktail (Sigma, P8340), and sonicated (Qsonica Q800 Sonicator) in polystyrene tubes at 45% power setting, 30 sec on/30 sec off for a total sonication time of 5 min at 4°C. The homogenate was centrifuged for 10 min at 16,000 g to remove debris, and the supernatant pre-cleared by incubation with 20 μL High Performance Streptavidin Sepharose Beads for 2 hr with rotation at 4°C.
Pre-cleared protein lysate was supplemented with 200 U/mL RNase Inhibitor (Roche) and added to RNA-bound beads for 2 hr with rotation at 4°C. Beads were washed 6 times with binding/lysis buffer supplemented with an additional 350 mM NaCl (500 mM final salt concentration). RNA-protein complexes were specifically eluted with 10 mM D-biotin (Thermo Fisher Scientific) in 50 μL binding/lysis buffer for 1 hr with rotation at 4°C. Eluate was concentrated by TCA precipitation, examined by denaturing RNA gel and SDS-PAGE with silver staining (Pierce Silver Stain Kit), and submitted for LC/MS protein identification (Taplan Mass Spectrometry Facility, Harvard Medical School).
Purification of His-tagged HNRNPK
6xHis-HNRNPK was purified from Rosetta-gami B bacteria as previously described (Tahir et al., 2014). Briefly, transformed bacteria were grown in Luria-Bertani broth at 37°C and 225 rpm to an optical density of 0.6 (600 nm), after which protein expression was induced for 4 hr at 25°C using 1 mM IPTG. Bacteria were harvested by centrifugation for 15 min at 4,000 rpm in a JA-14 rotor (Beckman), and resuspended in lysis buffer (50 mM Tris-HCl pH 8.0, 150 mM NaCl, 5% glycerol, 10 mM imidazole, 1x cOmplete EDTA-free protease inhibitor cocktail [Roche]). Bacteria were lysed by sonication (Branson Sonifier with attached microtip) using ten 10-sec pulses at power setting 6, while re-chilling lysate on ice in between pulses. Debris were pelleted by centrifugation for 15 min at 15,000 g at 4°C. The clarified lysate was combined with 0.5 mL Ni-NTA beads (Qiagen) pre-washed with lysis buffer, and incubated overnight at 4°C with rotation. The beads were transferred to a 10-mL polypropylene column and washed with 10 column volumes of wash buffer (20 mM Tris-HCl pH 6.8, 2 M NaCl, 60 mM imidazole, 1% Triton X-100). Protein was eluted from beads by incubation with 0.5 mL elution buffer (20 mM Tris-HCl pH 8.0, 5% glycerol, 500 mM imidazole, 5 μL. protease inhibitor cocktail [Sigma, P8340]) for 20 min at 4°C with rotation, and dialyzed against PBS containing 5% glycerol overnight at 4°C. The purity and concentration were analyzed by SDS-PAGE followed by Coomassie staining and Bradford assay (Bio-Rad Protein Assay Kit), respectively.
RNA EMSA
RNA EMSA was performed as previously described (Tomonaga and Levens, 1995). Briefly, RNA probes were generated by 5’-end-labeling with T4 polynucleotide kinase (New England BioLabs) and [γ-32P]ATP (PerkinElmer). Free nucleotides were removed using Illustra MicroSpin G-25 columns (GE Healthcare), and specific activity of probe was verified by scintillation counting to be ~500,000 cpm/μL. The following RNA oligos were used: WT Repeat B (GCCCCAGCCCCUGCCCCAGCCCCAGCCCCUGCCCCUGCCCCAGCCCCUGCCCCA); mutant Repeat B (GCAACAGCAACUGCAACAGCAACAGCAACUGCAACUGCAACAGCAACUGCAACA). Prior to performing EMSA, probe was denatured at 65°C for 5 min and folded by slow cooling to room temp. 50 fmol folded probe were added to 20 μL binding buffer (25 mM Tris-HCl pH 7.5, 200 mM glycine, 1 mM EDTA, 50 mM NaCl, 0.1% Tween-20, 10% glycerol, 0.5 mg/mL BSA, 2 μg/mL poly(dI-dC) [Thermo Fisher], 1 U/μL RNase inhibitor [Roche]) containing the indicated concentration of recombinant protein, and incubated for 30 min at room temp. Where indicated, unlabeled probe was added to binding reactions and pre-incubated for 10 min with protein prior to addition of labeled probe. RNA-protein complexes were resolved by electrophoresis on 1x TBE, 6% polyacrylamide gel at 4°C. The gel was drie d, exposed to storage phosphor screen (GE Healthcare), and imaged on Amersham Typhoon (GE Healthcare).
GFP-RYBP plasmid and HNRNPK siRNA delivery
RYBP cDNA was cloned into mammalian EGFP expression vector (derived from pSV2-EYFP-C1 (Tsukamoto et al., 2000)) and transfected into MEFs using Lipofectamine LTX (Thermo Fisher Scientific) as per manufacturer’s instructions. After 24 hr, cells were fixed and imaged by wide-field fluorescence microscopy. For siRNA knock-downs, 10 nM ON-TARGETplus SMARTpool targeting mouse HNRNPK (Dharmacon, L-048002-01) or non-targeting (scramble) control (Dharmacon, D-001810-10) was introduced into MEFs using Lipofectamine RNAiMAX (Thermo Fisher Scientific) as per manufacturer’s instructions. Knock-down was repeated daily for the indicated durations.
RT-PCR
RNA was isolated from cells using TRIzol Reagent (Thermo Fisher Scientific) as per manufacturer’s instructions. Genomic DNA was removed using TURBO DNase from the TURBO DNA-free Kit (Thermo Fisher Scientific). After inactivating TURBO DNase with DNase Inactivation Reagent (also enclosed in TURBO DNA-free Kit), DNAase-free RNA was reverse transcribed using SuperScript III Reverse Transcriptase (Thermo Fisher Scientific) with random primers (Promega, C118A) at 25°C for 5 min, 50°C fo r 1 hr, and 85°C for 15 min. Conventional PCR was performed on cDNA using 2x PCR MasterMix (Promega) or 2x Phusion High-Fidelity PCR Master Mix (New England BioLabs). Quantitative RT-PCR was performed using iTaq Universal SYBR Green Supermix (Bio-Rad) in a CFX96 Real-Time PCR Detection System (Bio-Rad). Gene-specific primer pairs are listed in Table S4.
ChIP-seq
ChIP-seq was performed as previously described (Minajigi et al., 2015), with minor modifications. Briefly, cells were cross-linked in PBS with 1% formaldehyde at room temp for 10 min with rotation at 1 million cells/mL, and quenched with 0.125 M glycine for 5 min. Cross-linked cells were washed twice with cold PBS, pelleted, and snap-frozen in liquid nitrogen. 10 million cross-linked cells per ChIP were thawed on ice and resuspended in 1 mL buffer 1 (50 nM HEPES pH 7.5, 140 mM NaCl, 1 mM EDTA, 0.5% NP-40, 0.25% Triton X-100, 10% glycerol, 1x cOmplete EDTA-free protease inhibitor cocktail [Roche]), and rotated for 10 min at 4°C. Nuclei were pelleted by centrifugation at 1,000 g for 5 min at 4°C, resuspended in 1 mL buffer 2 (10 mM HEPES pH 7.5, 200 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 1x cOmplete EDTA-free protease inhibitor cocktail) supplemented with 0.2 mg/mL RNase A (Thermo Fisher Scientific), and rotated for 10 min at 4°C. Nuclei were pelleted by centrifugation at 1,000 g for 5 min at 4°C and resuspended in 1.3 mL buffer 3 (10 mM HEPES pH 7.5, 100 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 1x cOmplete EDTA-free protease inhibitor cocktail, 0.1% sodium deoxycholate, 0.5% sarkosyl). Nuclei were sonicated (Qsonica Q800 Sonicator) in polystyrene tubes at 45% power reading, 30 sec on/30 sec off for a total sonication time of 4 min at 4°C. Triton X-100 was added to the lysate to 1%, which was then centrifuged for 10 min at 16,000 g to remove debris. The lysate was pre-cleared for 2 hr at 4°C with rotation using 20 μL Dynabeads Protein G (Thermo Fisher Scientific) pre-washed with PBS/0.5% BSA. After saving 10% as “input” sample, the pre-cleared lysate was combined with 20 mL Dynabeads Protein G pre-bound to 2 μg antibody (H3K27me3, GeneTex GTX60892; H2AK119ub, Cell Signaling CST8240), and rotated overnight at 4°C. Afterwards, beads were washed five times with wash buffer (50 mM HEPES pH 7.5, 0.5 M LiCl, 1 mM EDTA, 1% NP-40, 0.7% sodium deoxycholate), once with TEN buffer (10 mM Tris pH 8.0, 1 mM EDTA, 50 mM NaCl), and once with TE buffer (10 mM Tris pH 8.0, 1 mM EDTA). Input sample and beads containing ChIP material were resuspended in 400 μL TES buffer (50 mM Tris pH 8.0, 10 mM EDTA, 1% SDS) supplemented with 0.5 mg/mL Proteinase K (Sigma) and incubated for 1 hr at 55°C and then for >3 hr at 65°C to reverse cross-links. DNA was purified by phenol-chloroform extraction and quantified with Quant-iT PicoGreen dsDNA Reagent (Thermo Fisher Scientific). Input and ChIP-seq libraries were prepared in two biological replicates using NEBNext ChIP-seq Library Prep Master Mix Set for Illumina (New England BioLabs) as per manufacturer’s instructions. Libraries were sequenced on Illumina HiSeq2000 (high-throughput run) or HiSeq2500 (rapid run), generating ~50 million 50-nt paired-end reads per sample.
CHART-seq
Xist CHART-seq was performed as previously described (Wang et al., 2018). Briefly, cells were cross-linked in PBS with 1% formaldehyde at room temp for 10 min with rotation at 2 million cells/mL, and quenched with 0.125 M glycine for 5 min. Cross-linked cells were washed twice with cold PBS, pelleted, and snap-frozen in liquid nitrogen. 25 million cross-linked cells per CHART were thawed on ice and resuspended in 1 mL cold sucrose buffer (10 mM HEPES pH 7.5, 0.3 M sucrose, 1% Triton X-100, 100 mM potassium acetate, 0.1 mM EGTA) supplemented with 0.5 mM spermidine, 0.15 mM spermine, 1 mM DTT, 1x protease inhibitor cocktail (Sigma, P8340), 10 U/mL RNase inhibitor (Roche). The cell suspension was rotated for 10 min at 4°C, after which it was diluted with additional 2 mL cold sucrose buffer. This was added to a prechilled 15-mL glass Wheaton dounce tissue grinder (Thermo Fisher Scientific) that had been pre-rinsed once with RNaseZAP RNase decontamination solution (Thermo Fisher Scientific), three times with DEPC-treated water, and once with 2 mL sucrose buffer. Release of nuclei was accomplished by douncing cells 20 times with a tight pestle, and verified by staining with Trypan Blue Solution (Thermo Fisher Scientific). The nuclear suspension was then layered atop a cushion of 7.5 mL cold glycerol buffer (10 mM HEPES pH 7.5, 25% glycerol, 1 mM EDTA, 0.1 mM EGTA, 100 mM potassium acetate) supplemented with 0.5 mM spermidine, 0.15 mM spermine, 1 mM DTT, 1x cOmplete EDTA-free protease inhibitor cocktail (Roche), and 5 U/mL RNase inhibitor, and centrifuged at 1500 g for 10 min at 4°C. The purified nuclear pellet was resuspended in 6 mL PBS and further cross-linked with 3% formaldehyde for 30 min at room temp with rotation. Afterwards, nuclei were pelleted by centrifugation at 1,000 g for 5 min at 4°C and washed three times with cold PBS. Nuclei were resuspended in in 1 mL cold nuclear extraction buffer (50 mM HEPES pH 7.5, 250 mM NaCl, 0.1 mM EGTA, 0.5% sarkosyl, 0.1% sodium deoxycholate, 5 mM DTT, 10 U/mL RNase inhibitor) and incubated for 10 min at 4°C with rotation. Nuclei were then centrifuged at 400 g for 5 min at 4°C and resuspended in 230 μL cold sonication buffer (50 mM HEPES pH 7.5, 75 mM NaCl, 0.1 mM EGTA, 0.5% sarkosyl, 0.1% sodium deoxycholate, 0.1% SDS, 5 mM DTT, 10 U/mL RNase inhibitor) to a final volume of ~270 μL. Nuclei were sonicated for 5 min at 4°C using a C ovaris E220e (140W peak incident power, 10% duty factor, 200 cycles/burst). Sonicated chromatin was centrifuged at 16,000 g for 20 min at 4°C to remove debris. The supernatant (~220 μL) was mixed with 110 μL sonication buffer to a final volume of ~330 μL, which was split into two CHART reactions (Xist capture using sense probes and control capture using antisense probes).
For two CHART reactions, 600 μL MyOne Streptavidin C1 beads (Thermo Fisher Scientific) were used, which were pre-washed with 1 mL DEPC-treated water, blocked with 600 μL blocking buffer (33% sonication buffer, 67% 2x hybridization buffer, 500 ng/μL yeast RNA [Thermo Fisher Scientific], 1% BSA) at room temp for 1 hr, washed with 1 mL DEPC-treated water again, and resuspended in 610 μL 1x hybridization buffer (33% sonication buffer, 67% 2x hybridization buffer [see below for composition]). For each CHART, 160 μL chromatin was mixed with 320 μL 2x hybridization buffer (50 mM Tris pH 7.0, 750 mM NaCl, 1% SDS, 1 mM EDTA, 15% formamide, 1 mM DTT, 1 mM PMSF, 1x cOmplete EDTA-free protease inhibitor cocktail, 100 U/mL RNase inhibitor) and pre-cleared with 60 μL pre-blocked MyOne Streptavidin C1 beads at room temp for 1 hr with rotation. Pre-cleared chromatin was separated from beads, with 1% saved as “input” sample. The remaining pre-cleared chromatin (~500 μL) was mixed with 36 pmol of either antisense (Xist-targeting) or sense (control) biotinylated capture probes. We used a pool of 9 capture probes, which were selected from the 11 probes described previously (Simon et al., 2013), excluding probes X.3 and X.8. Hybridization was carried out at 37°C for 4 hr followed by incubation with 240 μL blocked MyOne Streptavidin C1 beads at 37°C for 1 hr. The beads were washed once with 1x hybridization buffer (33% sonication buffer, 67% 2x hybridization buffer) at 37°C for 10 min, five times with wash buffer (10 mM HEPES pH 7.5, 150 mM NaCl, 2% SDS, 2 mM EDTA, 2 mM EGTA, 1 mM DTT) at 37°C for 5 min, and twice with elution buffer (10 mM HEPES pH 7.5, 150 mM NaCl, 0.5% NP-40, 3 mM MgCl2, 10 mM DTT) at 37°C for 5 min. 10% of the final wash was saved as an “RNA capture” sample to estimate the efficiency of Xist RNA capture afterwards by RT-qPCR. CHART-enriched DNA was eluted twice in 200 μL elution buffer supplemented with 5 U/μL RNase H (New England BioLabs) at 37°C for 30 min. An “input” sample and CHART DNA were treated with 0.5 mg/mL RNase A (Thermo Fisher Scientific) at 37°C for 1 hr and then with 1% SDS, 10 mM EDTA, and 0.5 mg/mL proteinase K (Sigma) at 55°C for 1 hr. Reversal of crosslinks was performed by addition of 150 mM NaCl (final 300 mM) and incubation at 65°C overnight. DNA was purified by phenol-chloroform extraction and further sheared to <500-bp fragments using Covaris E220e (140W peak incident power, 10% duty factor, 200 cycles/burst). Sonicated DNA was purified using 1.8x Agencourt AMPure XP beads (Beckman Coulter). Input and CHART-seq libraries were prepared in two biological replicates using NEBNext ChIP-seq Library Prep Master Mix Set for Illumina (New England BioLabs) as per manufacturer’s instructions. Libraries were sequenced on Illumina HiSeq2000 (high-throughput run) or HiSeq2500 (rapid run), generating ~50 million 50-nt paired-end reads per sample.
ChIP-seq and CHART-seq analysis
To account for the hybrid character of our cell lines, adaptor-trimmed reads were separately aligned to custom mus/129 and cas genomes using NovoAlign (Novocraft), then mapped back to reference mm9 genome using SNPs (Pinter et al., 2012). This generated three tracks: composite of all reads (comp) and two allele-specific tracks using only allele-specifically mappable reads. We first used SPP (Kharchenko et al., 2008) to generate input-subtracted ChIP and CHART coverage profiles with smoothing using 1-kb windows recorded every 500 bp, as previously described (Simon et al., 2013). To account for different sequencing depths and CHART efficiencies, CHART profiles were scaled so that they have equal coverage within a 1-MB region flanking but not including the Xist locus (chrX:100150712-100650712 and 100683572-101183572). After scaling, we divided the coverage of each 1-kb window in ΔRepB, RING1A/B KO, and EED KO with that of WT, with a pseudo-count (0.001) introduced to each window. We then computed the base 2 logarithm of the ratio to generate the log2 ratio profiles. To account for different sequencing depths for ChIP-seq, samples differing by >10% were compensated by random downsampling with SAMtools (Li et al., 2009). To compare the densities of Xist, H3K27me3, and H2AK119ub at different categories of X-linked genes, we computed their densities over gene bodies by HOMER (Heinz et al., 2010). Non-expressed genes were defined as X-linked genes having zero RPKM in at least one RNA-seq replicate of WT MEFs; genes subject to XCI were defined as having non-zero RPKM in both WT MEF RNA-seq replicates after excluding escapees; escapees in the hybrid MEF line have been previously defined (Pinter et al., 2012). Genes with length shorter than 3 kb or overlapping unmappable regions were excluded from the analysis along with Xist and Tsix.
RNA-seq
Total cell RNA was extracted using TRIzol Reagent (Thermo Fisher Scientific), from which mRNA was isolated using NEBNext Poly(A) mRNA magnetic isolation module (New England BioLabs) as per manufacturer’s instructions. RNA-seq libraries were prepared in two biological replicates using NEBNext Ultra Directional RNA Library Prep Kit for Illumina (New England BioLabs) as per manufacturer’s instructions. Libraries were sequenced on Illumina HiSeq2000 (high-throughput run) or HiSeq2500 (rapid run), generating ~50 million 50-nt paired-end reads per sample.
RNA-seq analysis
PCR duplicates were removed and reads separately aligned to custom mus/129 and cas genomes using TopHat2 (Kim et al., 2013). Final allele-specific mapping to reference mm9 genome was generated based on SNPs (Pinter et al., 2012). Only uniquely aligned concordantly mapped sequences were used in downstream analysis. Counts per gene were calculated using featureCounts (Liao et al., 2014). Using MatLab (MathWorks), library sizes were normalized and genes with insufficient allelic information (<13 allele-specific reads) were removed. We also removed potentially miscalled genes from our alignment pipeline, defined as genes incorrectly assigned to mus from a pure cas RNA-seq library. Allele-specific RPKM was calculated using allelic ratio (allele-specific counts) applied to comp RPKM (total counts). For the CDF plot from fibroblast cells, genes with allele-specific RPKM≥1 were used while all other plots used genes with comp RPKM≥1 in at least one sample.
In situ Hi-C
In situ Hi-C was performed as previously described (Rao et al., 2014), with minor modifications outlined below. Briefly, 5 million cells per sample were cross-linked with 1% formaldehyde for 10 min at room temp. Cross-linked cells were lysed and the nuclei digested with 15 μL HindIII (100 U/μL; New England BioLabs, R0104M) at 37°C overnight. Cell clumps were removed by brief centrifugation, with the supernatant transferred to a new tube for the fill-in reaction with biotin-14-dATP (Thermo Fisher Scientific) and subsequent ligation. Re-ligated DNA was purified and then sheared to 300-500 bp in a Covaris microTUBE for 80 sec using Covaris E229e (140W peak incident power, 10% duty factor, 200 cycles/burst). Double size selection was performed on sheared DNA using 0.55x-0.75x Agencourt AMPure XP beads (Beckman Coulter). Biotinylated ligation products were purified with 30 μL MyOne Streptavidin C1 beads, end-repaired, dA-tailed, ligated to 3 μL 15 μM NEBNext Adaptor for Illumina (enclosed in NEBNext Multiplex Oligos for Illumina [Index Primers Set 1]; New England BioLabs), and digested with 3 μL USER enzyme (also enclosed in NEBNext Multiplex Oligos for Illumina [Primers Set 1]) at 37°C for 15 min. Hi-C libraries were then amplified with 12-16 cycles of PCR and cleaned up using Agencourt AMPure XP beads. Libraries were sequenced on Illumina HiSeq2000, generating ~200 million 50-nt paired-end reads per sample.
In situ Hi-C analysis
Reads were trimmed using cutadapt with the options --adapter=GATCGATC (MboI ligation junction) and --minimum-length=20. Reads of each pair were individually mapped to the mus (Xi) and cas (Xa) reference genomes using NovoAlign (Novocraft) and merged into Hi-C summary files and filtered using HOMER (Heinz et al., 2010) as previously described (Minajigi et al., 2015). Hi-C contact maps were generated from reads in the filtered HOMER mus and cas tag directories using the ‘pre’ command of Juicer tools with the genomeID mm9 (Durand et al., 2016b). The resulting Hi-C contact maps in .hic format were visualized and normalized with the ‘Balanced’ option in Juicebox (Durand et al., 2016a). Unless otherwise noted, all Hi-C interaction matrices in figures were visualized using Juicebox with 100-kb resolution and ‘Balanced’ normalization. Insulation scores, TADs, and TAD scores were determined using perl scripts previously described and available in the cworld GitHub repository (Giorgetti et al., 2016). Specifically, 100-kb resolution ‘Balanced’ normalized chromosome X Hi-C interaction matrices were extracted from Juicebox using the ‘dump’ function of Juicer tools (juicer dump observed KR name.hic X X BP 100000 name.KR.100kb.dump). Custom shell and R scripts were used to convert this output matrix into the cworld matrix format. Insulation scores were then calculated for the X chromosome using the resulting cworld matrix and the matrix2insulation.pl cworld script with parameters --is 500000 --ids 400000 --im iqrMean --nt 0 -- ss 200000 --yb 1.5 --nt 0 --bmoe 0. As described previously, regions of insulation score minima represent regions of locally high insulation and are considered potential TAD boundaries (Crane et al., 2015). TADs were detected from output insulation files using the cworld script insulation2tads.pl using default parameters (-- mbs 0 -- mts 0). To avoid slight differences in called TAD borders between WT and ΔRepB samples, and to avoid potential inaccuracies in the location of called TAD borders due to weakened TADs on the Xi, for each cell type (MEF or day 14 ES cell) TAD borders called on the WT Xa were used for further analysis of all samples. For example for MEF, TAD borders were called on the Xa and those borders used for analysis of Xi, ΔRepB Xa, and ΔRepB Xi MEF samples. For MEF Xa, 110 TADs were called while for day 14 ES cell, 108 TADs were called. TADs falling within low SNP density regions (33Mb-45Mb, 109116Mb, 123-130Mb) were excluded from further analysis, resulting in 89 TADs for MEF and 90 TADs for day 14 ES cell. The insulation2tads.pl script also calculated scores for each TAD. As previously described, the TAD score is calculated by first finding the maximum insulation score within a given TAD. The insulation scores at the boundaries of the TAD are then subtracted from this maximum insulation score. The TAD score is then taken to be the minimum of these two differences (https://github.com/dekkerlab/cworld-dekker). To determine if TAD scores overall differed significantly between samples, a Wilcoxon rank sum test was performed in R (wilcox.test(), paired=TRUE, all other options default). To determine the level of noise in the calculated TAD score between samples, for example due to differences in efficiency between Hi-C experiments, we first assumed that TAD strength should not differ substantially between the ΔRepB Xa and the WT Xa. Therefore, we calculated the ratio of TAD scores (ΔRepB/WT Xa) for each cell type (MEF or day 14 ES cell) and used this to assign an empirical false discovery rate (FDR) < 0.05 threshold for Xi. Only TADs with non-zero TAD scores on both the ΔRepB and WT Xa were included in the FDR < 0.05 threshold calculations. To determine which TADs were strengthened on ΔRepB versus WT Xi, ΔRepB/WT Xi TAD score ratios were calculated, and those greater than the FDR < 0.05 threshold were considered significantly strengthened. Analogously, TADs significantly weakened on ΔRepB versus WT Xi were also determined. For ΔRepB MEF Xi, 18 TADs were significantly strengthened and 7 significantly weakened. For ΔRepB day 14 ES cell Xi, 31 TADs were significantly strengthened and 8 were significantly weakened. To determine if X-linked genes that failed to be silenced in ΔRepB day 14 ES cells correlated with TADs that were significantly strengthened, fraction mus (Xi) reads (mus/[mus + cas]) were first calculated for each gene where allelic information could be determined (using the same criteria as above), in both the WT and ΔRepB day 14 RNA-seqs. The change in fraction mus reads (Δfraction mus reads) was then calculated for each gene (fraction ΔRepB mus reads/fraction WT mus reads). Each gene was then assigned to a day 14 ES cell TAD harboring the gene (with borders defined by the Xa, as explained above). For each gene, Δfraction mus reads were then plotted against the TAD score ratio (ΔRepB/WT ES Xi) of its assigned TAD. Linear correlation of RNA-seq ratios and TAD score ratios was then computed using the lm() function in R with default parameters. Principle component analysis (PCA) was done using the runHiCpca.pl command of HOMER with the filtered HOMER Hi-C tag directory as input and the options -res 100000 -superRes 100000 -genome mm9. Pearson correlation maps were also generated by HOMER using the analyzeHiC command with the filtered HOMER Hi-C tag directory as input and the options -chr chrX -res 200000 -corr. Pearson correlation maps were visualized with Java TreeView (Saldanha, 2004).
Quantification and Statistical Analysis
Statistical parameters including the statistical tests used and the values of n, p, and R are reported in the figures, figure legends, or associated main texts. Statistical significance is determined by the value of p < 0.05 by the indicated tests. For microscope images, n generally refers to the total number of counted cells or Xist clouds/Xi chromosomes.
Data and Software Availability
Original unprocessed gel, blot, and microscope images in this manuscript have been deposited at Mendeley Data and are available at: http://dx.doi.org/10.17632/cfk4bgz6w9.1
Raw high-throughput sequencing data and processed files for RNA-seq, ChIP-seq, CHART-seq, and Hi-C reported in this paper have been deposited at GEO under accession number: GSE107217
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
Rabbit monoclonal anti-H3K27me3 | GeneTex | Cat#GTX60892 |
Mouse monoclonal anti-H3K27me3 | Active Motif | Cat#AM39535 |
Mouse monoclonal anti-H3K27me3 | Cell Signaling | Cat#CST9733 |
Rabbit monoclonal anti-H2AK119ub | Cell Signaling | Cat#CST8240 |
Mouse monoclonal anti-EZH2 | Cell Signaling | Cat#CST3147 |
Rabbit polyclonal anti-HNRNPK | Proteintech | Cat#11426-1-AP |
Rabbit polyclonal anti-HNRNPK | Bethyl Labs | Cat#A300-674A |
Rabbit polyclonal anti-HNRNPE1 | Proteintech | Cat#14523-1-AP |
Rabbit polyclonal anti-HNRNPE2 | GeneTex | Cat#GTX114616 |
Rabbit polyclonal anti-HNRNPE3 | GeneTex | Cat#GTX118656 |
Rabbit monoclonal anti-GAPDH | Cell Signaling | Cat#CST14C10 |
Rabbit polyclonal anti-CIZ1 | Sunwoo et al., 2017 | N/A |
Chemicals, Peptides, and Recombinant Proteins | ||
Recombinant mouse HNRNPK | This study | N/A |
Recombinant mouse LIF | Sigma | Cat#ESG1107 |
KaryoMAX Colcemid Solution | Thermo Fisher Scientific | Cat#15212012 |
Critical Commercial Assays | ||
NEBNext Poly(A) mRNA Magnetic Isolation Module | New England BioLabs | Cat#E7490S |
Agencourt AMPure XP Beads | Beckman Coulter | Cat#A63881 |
NEBNext ChIP-Seq Library Prep Master Mix Set for Illumina | New England BioLabs | Cat#E6240S |
NEBNext Multiplex Oligos for Illumina (Index Primers Set 1) | New England BioLabs | Cat#E7335S |
Quant-iT PicoGreen dsDNA Reagent | Thermo Fisher Scientific | Cat#P7581 |
NEBNext Ultra Directional RNA Library Prep Kit for Illumina | New England BioLabs | Cat#E7420S |
Lipofectamine LTX with Plus Reagent | Thermo Fisher Scientific | Cat#15338100 |
Lipofectamine RNAiMAX Transfection Reagent | Thermo Fisher Scientific | Cat#13778150 |
Deposited Data | ||
Xist CHART-seq in WT female MEF | This study | GEO: GSE107217 |
Xist CHART-seq in ΔRepB Xi−/− female MEF | This study | GEO: GSE107217 |
H2AK119ub ChIP-seq in WT female MEF | This study | GEO: GSE107217 |
H2AK119ub ChIP-seq in ΔRepB Xi−/− female MEF | This study | GEO: GSE107217 |
H3K27me3 ChIP-seq in WT female MEF | This study | GEO: GSE107217 |
H3K27me3 ChIP-seq in ΔRepB Xi−/− female MEF | This study | GEO: GSE107217 |
Xist CHART-seq in Ring1A/B KO female MEF | This study | GEO: GSE107217 |
Xist CHART-seq in EED KO female MEF | This study | GEO: GSE107217 |
H3K27me3 ChIP-seq in RING1A/B KO female MEF (clone 13) | This study | GEO: GSE107217 |
H2AK119ub ChIP-seq in EED KO female MEF (clone 1-2) | This study | GEO: GSE107217 |
RNA-seq in WT female MEF | This study | GEO: GSE107217 |
RNA-seq in ΔRepB Xi−/− female MEF | This study | GEO: GSE107217 |
RNA-seq in WT female mouse differentiating ES cell (day 14) | This study | GEO: GSE107217 |
RNA-seq in ΔRepB female mouse differentiating ES cell (day 14) (clones C9 and D2) | This study | GEO: GSE107217 |
In situ Hi-C in WT female MEF | This study | GEO: GSE107217 |
In situ Hi-C in ΔRepB Xi−/− female MEF | This study | GEO: GSE107217 |
In situ Hi-C in WT female mouse differentiating ES cell (day 14) | This study | GEO: GSE107217 |
In situ Hi-C in ΔRepB female mouse differentiating ES cell (day 14) (clone D2) | This study | GEO: GSE107217 |
In situ Hi-C in WT female mouse differentiating ES cell (day 7) | Wang et al., 2018 | GEO: GSE99991 |
Mendeley data | This study | http://dx.doi.org/10.17632/cfk4bgz6w9.1 |
Experimental Models: Cell Lines | ||
Transgenic Xist exon 1 female MEF (♀X+P E1) | Jeon and Lee, 2011 | N/A |
Transgenic full-length Xist male MEF (♂X+P) | Jeon and Lee, 2011 | N/A |
WT Xist female MEF (EY.T4) | Yildirim et al., 2011 | N/A |
WT Xist female ES cell (TsixTST/+) | Ogawa et al., 2008 | N/A |
ΔRepA Xi+/− female MEF (clone X31) | This study | N/A |
ΔRepA Xi−/− female MEF (clone X9) | This study | N/A |
ΔRepF Xi+/− female MEF (clone 19) | This study | N/A |
ΔRepF Xi−/− female MEF (clone 6) | This study | N/A |
ΔRepB Xi+/− female MEF (clone 8) | This study | N/A |
ΔRepB Xi−/− female MEF (clone 22) | This study | N/A |
ΔRepC Xi+/− female MEF (clone 14) | This study | N/A |
ΔC-D Xi+/− female MEF (clone 22) | This study | N/A |
ΔRepD Xi+/− female MEF (clone 11) | This study | N/A |
ΔEx1 3' Xi+/− female MEF (clone 3) | This study | N/A |
ΔEx2-6 Xi+/− female MEF (clone 13) | This study | N/A |
ΔEx2-6 Xi−/− female MEF (clone H23) | This study | N/A |
ΔRepE Xi+/− female MEF (clone 4) | This study | N/A |
ΔRepE Xi−/− female MEF (clone 16) | This study | N/A |
ΔEx7a Xi+/− female MEF (clone N3) | This study | N/A |
ΔEx7b Xi+/− female MEF (clone H2) | This study | N/A |
ΔEx7c Xi+/− female MEF (clone 1) | This study | N/A |
ΔEx7d Xi+/− female MEF (clone N4) | This study | N/A |
ΔRepB Xa+Xi− female mouse ES cell (clone C9) | This study | N/A |
ΔRepB Xa−Xi− female mouse ES cell (clone D2) | This study | N/A |
EED KO female MEF (clone 1-2) | This study | N/A |
EED KO female MEF (clone 1-12) | This study | N/A |
RING1A/B KO female MEF (clone 67) | This study | N/A |
RING1A/B KO female MEF (clone 13) | This study | N/A |
Oligonucleotides | ||
ON-TARGETplus SMARTpool HNRNPK siRNA | Dharmacon | Cat#L-048002-01 |
ON-TARGETplus Non-targeting pool siRNA (scramble control) | Dharmacon | Cat#D-001810-10 |
gRNAs used to generate Xist deletions and HNRNPK, RING1A/B, and EED KOs (see Table S2) | Integrated DNA Technologies | N/A |
Oligo FISH probes used for Xist RNA FISH (see Table S3) | Integrated DNA Technologies | N/A |
XMP X Green Mouse Chromosome Paint | MetaSystems | Cat#D-1420-050-FI |
Biotinylated capture oligos used for Xist CHART | Simon et al., 2013 | N/A |
Probes used for RNA EMSA: WT Repeat B (GCCCCAGCCCCUGCCCCAGCCCCAGCCCCUGCCCCUGCCCCAGCCCCUGCCCCA); mutant Repeat B (GCAACAGCAACUGCAACAGCAACAGCAACUGCAACUGCAACAGCAACUGCAACA) | Integrated DNA Technologies | N/A |
Primers used for RT-qPCR (see Table S4) | Integrated DNA Technologies | N/A |
Primers used to generate Southern blot PCR probe: Primer 1 (CCTAAATGTCCTATAATCCATTGCTACACA); Primer 2 (CGCTTGGTGGATGGAAATATGGTTTTG) | Integrated DNA Technologies | N/A |
Recombinant DNA | ||
pSpCas9(BB)-2A-GFP (PX461) | Ran et al., 2013 | Addgene Cat#48140 |
pSpCas9(BB)-2A-Puro (PX459) v2.0 | Ran et al., 2013 | Addgene Cat#62988 |
EGFP mammalian expression vector (pSV2-EGFP-C1) | This study | N/A |
T7-4xS1m aptamer in vitro transcription vector (pDAC196) | This study | N/A |
Software and Algorithms | ||
HOMER v4.8 | Heinz et al., 2010 | http://homer.ucsd.edu/homer/index.html |
NovoAlign v3.02 | Novocraft | http://www.novocraft.com/products/novoalign/ |
matrix2insulation.pl | Giorgetti et al., 2016 | https://github.com/dekkerlab/giorgetti-nature-2016 |
insulation2tads.pl | Giorgetti et al., 2016 | https://github.com/dekkerlab/giorgetti-nature-2016 |
Juicebox v1.8.8 | Durand et al., 2016a | https://github.com/aidenlab/Juicebox/wiki/Download |
Juicer Tools | Durand et al., 2016b | https://github.com/aidenlab/juicer/wiki/Juicer-Tools-Quick-Start |
Java TreeView | Saldanha, 2004 | http://jtreeview.sourceforge.net |
TopHat2 v2.0.10 | Kim et al., 2013 | https://ccb.jhu.edu/software/tophat/index.shtml |
SPP | Kharchenko et al., 2008 | http://compbio.med.harvard.edu/Supplements/ChIP-seq/ |
featureCounts v1.5.0-p1 | Liao et al., 2014 | http://subread.sourceforge.net |
SAMtools v1.4.1 | Li et al., 2009 | http://samtools.sourceforge.net/ |
Supplementary Material
2
Table S1. List of proteins identified by LC/MS following in vitro RNA pulldown.
Related to Figure 3.
3
Table S2. gRNA target sequences used to generate Xist deletions and HNRNPK, RING1A/B, and EED KOs.
Related to STAR Methods.
4
Table S3. List of Xist oligo FISH probe sequences.
Related to STAR Methods.
5
Table S4. List of primer sequences used for quantitative RT-PCR.
Related to STAR Methods.
6
File S1. Sanger sequencing of Xist deletions and RING1A/B and EED KO clones.
Related to STAR Methods.
Both genomic DNA (gDNA) and cDNA were sequenced to verify: (1) genotypes correspond to Xi (Xist expressed) rather than Xa (Xist not expressed), and (2) appropriate splicing occurs. Where available, SNPs were also used to distinguish Xi (mus) and Xa (cas). Deleted regions are shown in red with gRNA target sites underlined. Introns and exons are shown in lower and upper case, respectively, with exon-exon junctions separated by a vertical bar (). Cryptic splice donor/acceptor sites are shown in bold, and insertion/point mutations are highlighted in yellow. mm9 coordinates for each deletion are provided.
ACKNOWLEDGMENTS
We thank all members of the Lee lab for critical comments and stimulating discussions. We acknowledge Hyun Jung Oh and Hsueh-Ping Chu for assistance with the CHART protocol, YongWoo Lee for help with preparation of mitotic chromosomes, Catherine Cifuentes-Rojas for characterization of EED KO clones, and John E. Froberg and Chunyao Wei for advice on bioinformatic analyses. H.S. was supported by NIH 5T32HD007396-24, A.K. by NSF GRFP and Herchel Smith Fellowship, and J.T.L. by NIH RO1-GM090278 and the Howard Hughes Medical Institute.
Footnotes
DECLARATION OF INTERESTS
J.T.L. is a co-founder and a member of the Scientific Advisory Boards of Translate Bio and Fulcrum Therapeutics.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- Almeida M, Pintacuda G, Masui O, Koseki Y, Gdula M, Cerase A, Brown D, Mould A, Innocent C, Nakayama M, et al. (2017). PCGF3/5-PRC1 initiates Polycomb recruitment in X chromosome inactivation. Science 356, 1081–1084 [Europe PMC free article] [Abstract] [Google Scholar]
- Bickmore WA, and van Steensel B (2013). Genome architecture: domain organization of interphase chromosomes. Cell 152, 1270–1284 [Abstract] [Google Scholar]
- Boettiger AN, Bintu B, Moffitt JR, Wang S, Beliveau BJ, Fudenberg G, Imakaev M, Mirny LA, Wu CT, and Zhuang X (2016). Super-resolution imaging reveals distinct chromatin folding for different epigenetic states. Nature 529, 418–422 [Europe PMC free article] [Abstract] [Google Scholar]
- Bomsztyk K, Denisenko O, and Ostrowski J (2004). hnRNP K: one protein multiple processes. Bioessays 26, 629–638 [Abstract] [Google Scholar]
- Bonev B, and Cavalli G (2016). Organization and function of the 3D genome. Nat Rev Genet 17, 661–678 [Abstract] [Google Scholar]
- Brown CJ, Hendrich BD, Rupert JL, Lafreniere RG, Xing Y, Lawrence J, and Willard HF (1992). The human XIST gene: analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell 71, 527–542 [Abstract] [Google Scholar]
- Brown CJ, and Willard HF (1994). The human X-inactivation centre is not required for maintenance of X-chromosome inactivation. Nature 368, 154–156 [Abstract] [Google Scholar]
- Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, and Zhang Y (2002). Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 298, 1039–1043 [Abstract] [Google Scholar]
- Chow JC, Hall LL, Baldry SE, Thorogood NP, Lawrence JB, and Brown CJ (2007). Inducible XIST-dependent X-chromosome inactivation in human somatic cells is reversible. Proc Natl Acad Sci U S A 104, 10104–10109 [Europe PMC free article] [Abstract] [Google Scholar]
- Chu C, Zhang QC, da Rocha ST, Flynn RA, Bharadwaj M, Calabrese JM, Magnuson T, Heard E, and Chang HY (2015). Systematic discovery of Xist RNA binding proteins. Cell 161, 404–416 [Europe PMC free article] [Abstract] [Google Scholar]
- Cirillo D, Blanco M, Armaos A, Buness A, Avner P, Guttman M, Cerase A, and Tartaglia GG (2016). Quantitative predictions of protein interactions with long noncoding RNAs. Nat Methods 14, 5–6 [Abstract] [Google Scholar]
- Clemson CM, McNeil JA, Willard HF, and Lawrence JB (1996). XIST RNA paints the inactive X chromosome at interphase: evidence for a novel RNA involved in nuclear/chromosome structure. J Cell Biol 132, 259–275 [Europe PMC free article] [Abstract] [Google Scholar]
- Cooper S, Grijzenhout A, Underwood E, Ancelin K, Zhang T, Nesterova TB, Anil-Kirmizitas B, Bassett A, Kooistra SM, Agger K, et al. (2016). Jarid2 binds mono-ubiquitylated H2A lysine 119 to mediate crosstalk between Polycomb complexes PRC1 and PRC2. Nat Commun 7, 13661. [Europe PMC free article] [Abstract] [Google Scholar]
- Crane E, Bian Q, McCord RP, Lajoie BR, Wheeler BS, Ralston EJ, Uzawa S, Dekker J, and Meyer BJ (2015). Condensin-driven remodelling of X chromosome topology during dosage compensation. Nature 523, 240–244 [Europe PMC free article] [Abstract] [Google Scholar]
- da Rocha ST, Boeva V, Escamilla-Del-Arenal M, Ancelin K, Granier C, Matias NR, Sanulli S, Chow J, Schulz E, Picard C, et al. (2014). Jarid2 Is Implicated in the Initial Xist-Induced Targeting of PRC2 to the Inactive X Chromosome. Mol Cell 53, 301–316 [Abstract] [Google Scholar]
- Darrow EM, Huntley MH, Dudchenko O, Stamenova EK, Durand NC, Sun Z, Huang SC, Sanborn AL, Machol I, Shamim M, et al. (2016). Deletion of DXZ4 on the human inactive X chromosome alters higher-order genome architecture. Proc Natl Acad Sci U S A 113, E4504–4512 [Europe PMC free article] [Abstract] [Google Scholar]
- Deng X, Ma W, Ramani V, Hill A, Yang F, Ay F, Berletch JB, Blau CA, Shendure J, Duan Z, et al. (2015). Bipartite structure of the inactive mouse X chromosome. Genome Biol 16, 152. [Europe PMC free article] [Abstract] [Google Scholar]
- Denisenko ON, and Bomsztyk K (1997). The product of the murine homolog of the Drosophila extra sex combs gene displays transcriptional repressor activity. Mol Cell Biol 17, 4707–4717 [Europe PMC free article] [Abstract] [Google Scholar]
- Disteche CM (2016). Dosage compensation of the sex chromosomes and autosomes. Semin Cell Dev Biol 56, 9–18 [Europe PMC free article] [Abstract] [Google Scholar]
- Dorafshan E, Kahn TG, and Schwartz YB (2017). Hierarchical recruitment of Polycomb complexes revisited. Nucleus 8, 496–505 [Europe PMC free article] [Abstract] [Google Scholar]
- Durand NC, Robinson JT, Shamim MS, Machol I, Mesirov JP, Lander ES, and Aiden EL (2016a). Juicebox Provides a Visualization System for Hi-C Contact Maps with Unlimited Zoom. Cell Syst 3, 99–101 [Europe PMC free article] [Abstract] [Google Scholar]
- Durand NC, Shamim MS, Machol I, Rao SS, Huntley MH, Lander ES, and Aiden EL (2016b). Juicer Provides a One-Click System for Analyzing Loop-Resolution Hi-C Experiments. Cell Syst 3, 95–98 [Europe PMC free article] [Abstract] [Google Scholar]
- Engreitz J, Pandya-Jones A, McDonel P, Shishkin A, Sirokman K, Surka C, Kadri S, Xing J, Goren A, Lander E, et al. (2013). The Xist lncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science (New York, NY) 341, 1237973 [Europe PMC free article] [Abstract] [Google Scholar]
- Francis NJ, Kingston RE, and Woodcock CL (2004). Chromatin compaction by a polycomb group protein complex. Science 306, 1574–1577 [Abstract] [Google Scholar]
- Froberg JE, Pinter SF, Kriz A, Jegu T, and Lee JT (2018). Megadomains and superloops form dynamically but are dispensable for X-chromosome inactivation and gene escape. Nat Commun in press [Europe PMC free article] [Abstract] [Google Scholar]
- Gallardo M, Lee HJ, Zhang X, Bueso-Ramos C, Pageon LR, McArthur M, Multani A, Nazha A, Manshouri T, Parker-Thornburg J, et al. (2015). hnRNP K Is a Haploinsufficient Tumor Suppressor that Regulates Proliferation and Differentiation Programs in Hematologic Malignancies. Cancer Cell 28, 486–499 [Europe PMC free article] [Abstract] [Google Scholar]
- Giorgetti L, Lajoie BR, Carter AC, Attia M, Zhan Y, Xu J, Chen CJ, Kaplan N, Chang HY, Heard E, et al. (2016). Structural organization of the inactive X chromosome in the mouse. Nature 535, 575–579 [Europe PMC free article] [Abstract] [Google Scholar]
- Hasegawa Y, Brockdorff N, Kawano S, Tsutui K, Tsutui K, and Nakagawa S (2010). The matrix protein hnRNP U is required for chromosomal localization of Xist RNA. Dev Cell 19, 469–476 [Abstract] [Google Scholar]
- Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, Cheng JX, Murre C, Singh H, and Glass CK (2010). Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38, 576–589 [Europe PMC free article] [Abstract] [Google Scholar]
- Hoki Y, Kimura N, Kanbayashi M, Amakawa Y, Ohhata T, Sasaki H, and Sado T (2009). A proximal conserved repeat in the Xist gene is essential as a genomic element for X-inactivation in mouse. Development 136, 139–146 [Abstract] [Google Scholar]
- Jeon Y, and Lee JT (2011). YY1 tethers Xist RNA to the inactive X nucleation center. Cell 146, 119–133 [Europe PMC free article] [Abstract] [Google Scholar]
- Kahn TG, Dorafshan E, Schultheis D, Zare A, Stenberg P, Reim I, Pirrotta V, and Schwartz YB (2016). Interdependence of PRC1 and PRC2 for recruitment to Polycomb Response Elements. Nucleic Acids Res 44, 10132–10149 [Europe PMC free article] [Abstract] [Google Scholar]
- Kalb R, Latwiel S, Baymaz HI, Jansen PW, Muller CW, Vermeulen M, and Muller J (2014). Histone H2A monoubiquitination promotes histone H3 methylation in Polycomb repression. Nat Struct Mol Biol 21, 569–571 [Abstract] [Google Scholar]
- Kharchenko PV, Tolstorukov MY, and Park PJ (2008). Design and analysis of ChIP-seq experiments for DNA-binding proteins. Nat Biotechnol 26, 1351–1359 [Europe PMC free article] [Abstract] [Google Scholar]
- Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, and Salzberg SL (2013). TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol 14, R36. [Europe PMC free article] [Abstract] [Google Scholar]
- Kundu S, Ji F, Sunwoo H, Jain G, Lee JT, Sadreyev RI, Dekker J, and Kingston RE (2018). Polycomb Repressive Complex 1 Generates Discrete Compacted Domains that Change during Differentiation. Mol Cell 71, 191. [Europe PMC free article] [Abstract] [Google Scholar]
- Lee JT (2011). Gracefully ageing at 50, X-chromosome inactivation becomes a paradigm for RNA and chromatin control. Nat Rev Mol Cell Biol 12, 815–826 [Abstract] [Google Scholar]
- Lee JT, Lu N, and Han Y (1999). Genetic analysis of the mouse X inactivation center defines an 80-kb multifunction domain. Proc Natl Acad Sci U S A 96, 3836–3841 [Europe PMC free article] [Abstract] [Google Scholar]
- Leppek K, and Stoecklin G (2014). An optimized streptavidin-binding RNA aptamer for purification of ribonucleoprotein complexes identifies novel ARE-binding proteins. Nucleic Acids Res 42, e13. [Europe PMC free article] [Abstract] [Google Scholar]
- Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, and Genome Project Data Processing, S. (2009). The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 [Europe PMC free article] [Abstract] [Google Scholar]
- Liao Y, Smyth GK, and Shi W (2014). featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 [Abstract] [Google Scholar]
- Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A, Amit I, Lajoie BR, Sabo PJ, Dorschner MO, et al. (2009). Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 [Europe PMC free article] [Abstract] [Google Scholar]
- Makhlouf M, Ouimette JF, Oldfield A, Navarro P, Neuillet D, and Rougeulle C (2014). A prominent and conserved role for YY1 in Xist transcriptional activation. Nat Commun 5, 4878. [Europe PMC free article] [Abstract] [Google Scholar]
- Marahrens Y, Panning B, Dausman J, Strauss W, and Jaenisch R (1997). Xist-deficient mice are defective in dosage compensation but not spermatogenesis. Genes Dev 11, 156–166 [Abstract] [Google Scholar]
- Margueron R, and Reinberg D (2011). The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 [Europe PMC free article] [Abstract] [Google Scholar]
- McHugh CA, Chen CK, Chow A, Surka CF, Tran C, McDonel P, Pandya-Jones A, Blanco M, Burghard C, Moradian A, et al. (2015). The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature 521, 232–236 [Europe PMC free article] [Abstract] [Google Scholar]
- Minajigi A, Froberg J, Wei C, Sunwoo H, Kesner B, Colognori D, Lessing D, Payer B, Boukhali M, Haas W, et al. (2015). Chromosomes. A comprehensive Xist interactome reveals cohesin repulsion and an RNA-directed chromosome conformation. Science 349 [Europe PMC free article] [Abstract] [Google Scholar]
- Mira-Bontenbal H, and Gribnau J (2016). New Xist-Interacting Proteins in X-Chromosome Inactivation. Curr Biol 26, R338–342 [Abstract] [Google Scholar]
- Moindrot B, Cerase A, Coker H, Masui O, Grijzenhout A, Pintacuda G, Schermelleh L, Nesterova TB, and Brockdorff N (2015). A Pooled shRNA Screen Identifies Rbm15, Spen, and Wtap as Factors Required for Xist RNA-Mediated Silencing. Cell Rep 12, 562–572 [Europe PMC free article] [Abstract] [Google Scholar]
- Monfort A, Di Minin G, Postlmayr A, Freimann R, Arieti F, Thore S, and Wutz A (2015). Identification of Spen as a Crucial Factor for Xist Function through Forward Genetic Screening in Haploid Embryonic Stem Cells. Cell Rep 12, 554–561 [Europe PMC free article] [Abstract] [Google Scholar]
- Nora EP, Lajoie BR, Schulz EG, Giorgetti L, Okamoto I, Servant N, Piolot T, van Berkum NL, Meisig J, Sedat J, et al. (2012). Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 [Europe PMC free article] [Abstract] [Google Scholar]
- Ogawa Y, Sun BK, and Lee JT (2008). Intersection of the RNA interference and X-inactivation pathways. Science 320, 1336–1341 [Europe PMC free article] [Abstract] [Google Scholar]
- Oksuz O, Narendra V, Lee CH, Descostes N, LeRoy G, Raviram R, Blumenberg L, Karch K, Rocha PP, Garcia BA, et al. (2018). Capturing the Onset of PRC2-Mediated Repressive Domain Formation. Mol Cell 70, 1149–1162 e1145 [Europe PMC free article] [Abstract] [Google Scholar]
- Pintacuda G, Wei G, Roustan C, Kirmizitas BA, Solcan N, Cerase A, Castello A, Mohammed S, Moindrot B, Nesterova TB, et al. (2017). hnRNPK Recruits PCGF3/5-PRC1 to the Xist RNA B-Repeat to Establish Polycomb-Mediated Chromosomal Silencing. Mol Cell 68, 955–969 e910 [Europe PMC free article] [Abstract] [Google Scholar]
- Pinter SF, Sadreyev RI, Yildirim E, Jeon Y, Ohsumi TK, Borowsky M, and Lee JT (2012). Spreading of X chromosome inactivation via a hierarchy of defined Polycomb stations. Genome Res 22, 1864–1876 [Europe PMC free article] [Abstract] [Google Scholar]
- Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, and Zhang F (2013). Genome engineering using the CRISPR-Cas9 system. Nat Protoc 8, 2281–2308 [Europe PMC free article] [Abstract] [Google Scholar]
- Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, Sanborn AL, Machol I, Omer AD, Lander ES, et al. (2014). A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 [Europe PMC free article] [Abstract] [Google Scholar]
- Ridings-Figueroa R, Stewart ER, Nesterova TB, Coker H, Pintacuda G, Godwin J, Wilson R, Haslam A, Lilley F, Ruigrok R, et al. (2017). The nuclear matrix protein CIZ1 facilitates localization of Xist RNA to the inactive X-chromosome territory. Genes Dev 31, 876–888 [Europe PMC free article] [Abstract] [Google Scholar]
- Royce-Tolland ME, Andersen AA, Koyfman HR, Talbot DJ, Wutz A, Tonks ID, Kay GF, and Panning B (2010). The A-repeat links ASF/SF2-dependent Xist RNA processing with random choice during X inactivation. Nat Struct Mol Biol 17, 948–954 [Europe PMC free article] [Abstract] [Google Scholar]
- Saldanha AJ (2004). Java Treeview--extensible visualization of microarray data. Bioinformatics 20, 3246–3248 [Abstract] [Google Scholar]
- Schoeftner S, Sengupta AK, Kubicek S, Mechtler K, Spahn L, Koseki H, Jenuwein T, and Wutz A (2006). Recruitment of PRC1 function at the initiation of X inactivation independent of PRC2 and silencing. EMBO J 25, 3110–3122 [Europe PMC free article] [Abstract] [Google Scholar]
- Simon JA, and Kingston RE (2013). Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol Cell 49, 808–824 [Europe PMC free article] [Abstract] [Google Scholar]
- Simon MD, Pinter SF, Fang R, Sarma K, Rutenberg-Schoenberg M, Bowman SK, Kesner BA, Maier VK, Kingston RE, and Lee JT (2013). High-resolution Xist binding maps reveal two-step spreading during X-chromosome inactivation. Nature 504, 465–469 [Europe PMC free article] [Abstract] [Google Scholar]
- Srisawat C, and Engelke DR (2001). Streptavidin aptamers: affinity tags for the study of RNAs and ribonucleoproteins. RNA 7, 632–641 [Europe PMC free article] [Abstract] [Google Scholar]
- Starmer J, and Magnuson T (2009). A new model for random X chromosome inactivation. Development 136, 1–10 [Europe PMC free article] [Abstract] [Google Scholar]
- Sunwoo H, Colognori D, Froberg JE, Jeon Y, and Lee JT (2017). Repeat E anchors Xist RNA to the inactive X chromosomal compartment through CDKN1A-interacting protein (CIZ1). Proc Natl Acad Sci U S A [Europe PMC free article] [Abstract] [Google Scholar]
- Sunwoo H, Wu JY, and Lee JT (2015). The Xist RNA-PRC2 complex at 20-nm resolution reveals a low Xist stoichiometry and suggests a hit-and-run mechanism in mouse cells. Proc Natl Acad Sci U S A 112, E4216–4225 [Europe PMC free article] [Abstract] [Google Scholar]
- Tahir TA, Singh H, and Brindle NP (2014). The RNA binding protein hnRNP-K mediates post-transcriptional regulation of uncoupling protein-2 by angiopoietin-1. Cell Signal 26, 1379–1384 [Europe PMC free article] [Abstract] [Google Scholar]
- Tavares L, Dimitrova E, Oxley D, Webster J, Poot R, Demmers J, Bezstarosti K, Taylor S, Ura H, Koide H, et al. (2012). RYBP-PRC1 complexes mediate H2A ubiquitylation at polycomb target sites independently of PRC2 and H3K27me3. Cell 148, 664–678 [Europe PMC free article] [Abstract] [Google Scholar]
- Tomonaga T, and Levens D (1995). Heterogeneous nuclear ribonucleoprotein K is a DNA-binding transactivator. J Biol Chem 270, 4875–4881 [Abstract] [Google Scholar]
- Tsukamoto T, Hashiguchi N, Janicki SM, Tumbar T, Belmont AS, and Spector DL (2000). Visualization of gene activity in living cells. Nat Cell Biol 2, 871–878 [Abstract] [Google Scholar]
- Wang CY, Jegu T, Chu HP, Oh HJ, and Lee JT (2018). SMCHD1 Merges Chromosome Compartments and Assists Formation of Super-Structures on the Inactive X. Cell [Europe PMC free article] [Abstract] [Google Scholar]
- Wutz A, Rasmussen TP, and Jaenisch R (2002). Chromosomal silencing and localization are mediated by different domains of Xist RNA. Nat Genet 30, 167–174 [Abstract] [Google Scholar]
- Yildirim E, Kirby JE, Brown DE, Mercier FE, Sadreyev RI, Scadden DT, and Lee JT (2013). Xist RNA is a potent suppressor of hematologic cancer in mice. Cell 152, 727–742 [Europe PMC free article] [Abstract] [Google Scholar]
- Yildirim E, Sadreyev RI, Pinter SF, and Lee JT (2011). X-chromosome hyperactivation in mammals via nonlinear relationships between chromatin states and transcription. Nat Struct Mol Biol 19, 56–61 [Europe PMC free article] [Abstract] [Google Scholar]
- Yue M, Ogawa A, Yamada N, Charles Richard JL, Barski A, and Ogawa Y (2017). Xist RNA repeat E is essential for ASH2L recruitment to the inactive X and regulates histone modifications and escape gene expression. PLoS Genet 13, e1006890. [Europe PMC free article] [Abstract] [Google Scholar]
- Zhao J, Sun BK, Erwin JA, Song JJ, and Lee JT (2008). Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 322, 750–756 [Europe PMC free article] [Abstract] [Google Scholar]
Full text links
Read article at publisher's site: https://doi.org/10.1016/j.molcel.2019.01.015
Read article for free, from open access legal sources, via Unpaywall: http://www.cell.com/article/S1097276519300358/pdf
Citations & impact
Impact metrics
Citations of article over time
Alternative metrics
Smart citations by scite.ai
Explore citation contexts and check if this article has been
supported or disputed.
https://scite.ai/reports/10.1016/j.molcel.2019.01.015
Article citations
HOXDeRNA activates a cancerous transcription program and super enhancers via genome-wide binding.
Mol Cell, 84(20):3950-3966.e6, 08 Oct 2024
Cited by: 1 article | PMID: 39383879
Female-bias in systemic lupus erythematosus: How much is the X chromosome to blame?
Biol Sex Differ, 15(1):76, 07 Oct 2024
Cited by: 0 articles | PMID: 39375734 | PMCID: PMC11460073
Review Free full text in Europe PMC
Multifaceted role of CTCF in X-chromosome inactivation.
Chromosoma, 133(4):217-231, 21 Oct 2024
Cited by: 0 articles | PMID: 39433641
Review
lncRNA HOTAIR and Cardiovascular diseases.
Funct Integr Genomics, 24(5):165, 19 Sep 2024
Cited by: 0 articles | PMID: 39294422
Review
A critical role for X-chromosome architecture in mammalian X-chromosome dosage compensation.
Curr Opin Genet Dev, 87:102235, 25 Jul 2024
Cited by: 0 articles | PMID: 39053028
Review
Go to all (92) article citations
Data
Data behind the article
This data has been text mined from the article, or deposited into data resources.
BioStudies: supplemental material and supporting data
Data Citations
- (2 citations) DOI - 10.17632/cfk4bgz6w9.1
GEO - Gene Expression Omnibus (2)
- (19 citations) GEO - GSE107217
- (1 citation) GEO - GSE99991
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
hnRNPK Recruits PCGF3/5-PRC1 to the Xist RNA B-Repeat to Establish Polycomb-Mediated Chromosomal Silencing.
Mol Cell, 68(5):955-969.e10, 01 Dec 2017
Cited by: 166 articles | PMID: 29220657 | PMCID: PMC5735038
G-quadruplex folding in Xist RNA antagonizes PRC2 activity for stepwise regulation of X chromosome inactivation.
Mol Cell, 84(10):1870-1885.e9, 01 May 2024
Cited by: 1 article | PMID: 38759625
The Xist RNA-PRC2 complex at 20-nm resolution reveals a low Xist stoichiometry and suggests a hit-and-run mechanism in mouse cells.
Proc Natl Acad Sci U S A, 112(31):E4216-25, 20 Jul 2015
Cited by: 63 articles | PMID: 26195790 | PMCID: PMC4534268
The many faces of Polycomb regulation by RNA.
Curr Opin Genet Dev, 61:53-61, 01 Apr 2020
Cited by: 28 articles | PMID: 32403014 | PMCID: PMC7653676
Review Free full text in Europe PMC
Funding
Funders who supported this work.
Herchel Smith Fellowship
Howard Hughes Medical Institute
NICHD NIH HHS (2)
Grant ID: T32 HD007396
Grant ID: R01 HD097665
NIGMS NIH HHS (1)
Grant ID: R01 GM090278
NIH (2)
Grant ID: 5T32HD007396-24
Grant ID: RO1-GM090278