Europe PMC
Nothing Special   »   [go: up one dir, main page]

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


The specificity of gene expression during development requires the insulation of regulatory domains to avoid inappropriate enhancer-gene interactions. In vertebrates, this insulator function is mostly attributed to clusters of CTCF sites located at topologically associating domain (TAD) boundaries. However, TAD boundaries allow some physical crosstalk across regulatory domains, which is at odds with the specific and precise expression of developmental genes. Here we show that developmental genes and nearby clusters of CTCF sites cooperatively foster the robust insulation of regulatory domains. By genetically dissecting a couple of representative loci in mouse embryonic stem cells, we show that CTCF sites prevent undesirable enhancer-gene contacts (i.e. physical insulation), while developmental genes preferentially contribute to regulatory insulation through non-structural mechanisms involving promoter competition rather than enhancer blocking. Overall, our work provides important insights into the insulation of regulatory domains, which in turn might help interpreting the pathological consequences of certain structural variants.

Free full text 


Logo of ncommsLink to Publisher's site
Nat Commun. 2024; 15: 7258.
Published online 2024 Aug 23. https://doi.org/10.1038/s41467-024-51602-4
PMCID: PMC11344162
PMID: 39179577

Cooperative insulation of regulatory domains by CTCF-dependent physical insulation and promoter competition

Associated Data

Supplementary Materials
Data Availability Statement

Abstract

The specificity of gene expression during development requires the insulation of regulatory domains to avoid inappropriate enhancer-gene interactions. In vertebrates, this insulator function is mostly attributed to clusters of CTCF sites located at topologically associating domain (TAD) boundaries. However, TAD boundaries allow some physical crosstalk across regulatory domains, which is at odds with the specific and precise expression of developmental genes. Here we show that developmental genes and nearby clusters of CTCF sites cooperatively foster the robust insulation of regulatory domains. By genetically dissecting a couple of representative loci in mouse embryonic stem cells, we show that CTCF sites prevent undesirable enhancer-gene contacts (i.e. physical insulation), while developmental genes preferentially contribute to regulatory insulation through non-structural mechanisms involving promoter competition rather than enhancer blocking. Overall, our work provides important insights into the insulation of regulatory domains, which in turn might help interpreting the pathological consequences of certain structural variants.

Subject terms: Gene regulation, Gene expression, Functional genomics

Introduction

The specific and precise expression of developmental genes during embryogenesis requires the concerted action of several types of cis-regulatory elements1,2. Among them, enhancers are able to activate gene transcription by communicating with gene promoters across large linear distances1,3. In contrast, insulators protect gene promoters from signals emanating from neighboring regulatory domains by either blocking the communication with non-cognate enhancers (i.e. enhancer blocking insulators) or by acting as barriers against the spreading of repressive chromatin (i.e. boundary elements)4,5. In order to execute their enhancer-blocking function, insulators need to be located between an enhancer and the protected promoter. Thus, enhancer activity is typically constrained within discrete regulatory domains containing cognate gene promoters and demarcated by insulators at both ends4,6,7. Insulators are best understood in Drosophila, where the combinatorial binding of several architectural proteins (i.e. Cp190, CTCF, Su(Hw), BEAF-32, GAF) to these elements enables their enhancer-blocking activity810. In mammals, the repertoire of architectural proteins is markedly reduced and CTCF is considered the main insulator-binding protein6,11,12

More recently, the emergence of novel methodologies, such as Hi-C, has resolved the 3D organization of genomes at high resolution. These studies revealed that, in mammals, CTCF sites often coincide with the boundaries of large self-interacting genomic regions that were termed topologically associating domains (TADs)13. Notably, TADs often overlap with regulatory domains, particularly those containing major developmental genes whose expression is regulated by long-range enhancers14. Furthermore, these developmental genes are often located within TADs whose boundaries display particularly strong evolutionary conservation14. In mammals, the formation of TAD boundaries involves the stalling of loop-extruding cohesin complexes by CTCF11,15,16. In contrast, cohesin and loop extrusion might not play a major role in TAD boundary formation in Drosophila, which might instead involve pairwise looping between insulator elements due to the dimerization of architectural proteins10,17,18. Furthermore, although architectural proteins play a preponderant role in TAD boundary formation, with, for example, over 80% of mammalian TAD boundaries being CTCF dependent, they seem to be dispensable for or even counteract the organization of chromatin compartments11,15,16. Interestingly, TAD boundaries that are not dependent on CTCF are often located proximal to active transcription start sites (TSS) and housekeeping genes13,19. Recent reports indicate that RNA Pol2 can participate in the formation of contact domains and physical boundaries2026, but whether this can globally contribute to the insulation of regulatory domains and enhancer-blocking remains controversial2729. In fact, due to the intermingling of chromatin domains, it has been shown that, at least at certain loci, the location of genes near or within boundaries might facilitate, rather than block, the communication with enhancers located along neighbouring TADs3032. Similarly, strong enhancers might be able to bypass boundaries and activate genes across TADs33,34. These observations suggest that, rather than impenetrable walls, TAD boundaries might act as dynamic and partially permeable barriers, allowing certain level of physical crosstalk across regulatory domains35,36. On the other hand, in mammals, the CTCF clusters that often overlap with TAD boundaries might not only act as insulators but also as tethering elements that facilitate the physical communication of nearby genes with distal enhancers located within the same TAD3741. Overall, TAD boundaries seem to play a dual regulatory role, as they can facilitate enhancer-gene communication within TADs while preventing, albeit partly, undesired enhancer-gene contacts across TADs42.

The permeability of TAD boundaries is at odds with the remarkable tissue specificity and spatiotemporal precision with which most developmental genes are expressed, even in the absence of CTCF11,18 or other architectural proteins8,10,15. Therefore, besides boundary elements, additional mechanisms might ensure the robust insulation of developmental regulatory domains. In this regard, work mostly based on the genetic dissection of the mammalian alpha and beta globin loci, as well as reporter assays in Drosophila, suggests that promoters can also regulate enhancer-gene communication through either promoter competition or enhancer blocking24,4350. When located within the same domain, several promoters can, in principle, share and get activated by a common enhancer5153. However, depending on the presence of distinct promoter elements, the activation of a preferred gene can preclude the expression of its neighbours independently of insulators. Promoter competition occurs when the preferred gene gets specifically activated regardless of its relative position with respect to the shared enhancer/s and neighbouring gene/s47,54. It has been previously suggested that the competition between promoters for a shared enhancer might involve mutually exclusive enhancer-promoter contacts (flip-flop model)44,55. In addition, recent observations suggest that, at certain loci, promoter competition could also entail non-structural mechanisms whereby promoters and enhancers share transcriptional hubs/condensates51,52, within which promoters might compete for rate-limiting factors (e.g. TFs, GTFs, RNA Pol2) required for gene transcription56,57. In contrast, promoter-driven enhancer blocking occurs when the preferred gene prevents the expression of its neighbours only if placed between the shared enhancer/s and the other gene/s, thus resembling how insulators work24,48. Recent studies suggest that promoter-driven enhancer blocking might involve structural mechanisms, whereby protein complexes present at promoters (e.g. RNA Pol2) act as weak barriers against cohesin-mediated loop extrusion2123,25. However, other studies based on the depletion or inhibition of RNA Pol2 have reported small or no effects in 3D chromatin architecture and formation of TAD boundaries2729. Overall, it is currently unclear how prevalent these two promoter-dependent mechanisms are within endogenous loci and whether they significantly contribute to gene expression specificity, particularly in mammals. Similarly, it is largely unknown whether insulators, architectural proteins and promoters might somehow crosstalk in order to modulate the insulation of regulatory domains26,49.

In the present study, we uncovered that a significant fraction of developmental genes in both mice and humans are located near TAD boundaries that contain clusters of CTCF binding sites. Furthermore, at these boundaries, developmental genes and CTCF sites are often organized in a sequential and evolutionary conserved manner, with the genes preceding the CTCF clusters. In contrast, genes previously reported as capable of bypassing boundaries3032 are typically flanked by CTCF sites and, thus, located within CTFC clusters. Most importantly, through the exhaustive genetic dissection of a couple of representative developmental loci (i.e. Gbx2/Asb18, Six3/Six2), we show that the positioning of developmental genes close to boundaries does not facilitate their own expression. Instead, we found that developmental genes and CTCF sites strengthen the regulatory insulation capacity of the nearby TAD boundaries. Finally, we show that developmental genes seem to preferentially contribute to regulatory insulation through non-structural mechanisms that involve promoter competition rather than enhancer blocking. Overall, our work provides important insights into the mechanisms contributing to the robust and specific expression of developmental genes during mammalian embryogenesis.

Results

TADs containing developmental genes display distinctive features

While dissecting the regulatory landscapes of a few representative developmental genes characterized by the presence of large CpG island (CGI) clusters and Polycomb-Group (PcG) protein domains at their promoters (e.g. TFAPA, Six3, Lhx5)58,59, we noticed that these genes were often located within TADs displaying rather unique features. Namely, these developmental TADs (i) showed low gene density and (ii) displayed a skewed gene distribution, as the developmental genes were often located near TAD boundaries (Figs. S12). To evaluate whether these features are prevalent among developmental TADs, we used TAD maps previously generated in either mouse or human cells and defined developmental genes based on the presence of broad PcG domains around their TSS (See Methods)6062. This definition does not include all developmental genes according to Gene Ontology (GO) terms, but rather selects a subset of major developmental genes whose promoters include large CGI clusters and display strong enhancer responsiveness6365. Nevertheless, for the sake of simplicity, and unless stated otherwise, we will refer to this subset of genes with broad PcG domains around their TSS as developmental genes. Using this definition of developmental genes, the following analyses were performed:

(i) Gene density: We first classified TADs previously identified in mESC and hESC according to gene density (High, Medium or Low) and performed Gene Ontology analyses for the genes contained within each TAD category. Interestingly, TADs with low gene density were strongly and specifically enriched in developmental genes, while High and Medium gene density TADs showed milder gene ontology enrichments and were not particularly enriched in developmental terms (Fig. 1A, Fig. S3, Data S12). Next, to evaluate whether these observations could be generalized, we considered TAD maps previously generated in additional human (n = 37) and mouse (n = 11) cell types. Notably, developmental genes (i.e. genes with broad PcG domains) were significantly enriched within TADs with low gene density in all the considered mouse and human cell types (Fisher test p-values obtained were smaller than 4.2e-05 and 1.6e-10 in humans and mice, respectively, for all tests performed, with an average odds ratio of 2.0 (95% confidence interval: 1.9 to 2.1) in humans and 2.1 in mice (95% confidence interval: 2.0 to 2.2). In contrast, housekeeping genes were not enriched within low gene density TADs in any of the analyzed TAD maps (Fisher test p-value = 1 for all tests, average odds ratio of 0.36 and 0.65 in humans and mice, respectively).

An external file that holds a picture, illustration, etc.
Object name is 41467_2024_51602_Fig1_HTML.jpg
TADs containing developmental genes have distinctive features.

A TADs previously identified in mESC13 were classified based on their gene density in three different groups: High Density (HD), Medium Density (MD) and Low Density (LD). Then, the genes present within each TAD group were subject to GO enrichment analysis. For the MD (n = 17 enriched GO terms) and LD (n = 135 enriched GO terms) groups, only the top 10 most significantly enriched GO terms are highlighted (i.e. Q value  0.05), while for the HD group all the significantly enriched GO terms (n = 8) are presented. B The distribution of different groups of genes within TADs was investigated using previously generated TAD maps. TADs were divided in 10 bins of equal sizes, which were then grouped in five bin pairs based on their distance to the nearest boundary (e.g. Bin 1 is the closest to TAD boundaries; Bin 5 is the most distal from TAD boundaries) (see Methods). Bin 0 indicates genes located at inter-TAD regions (i.e. within TAD boundaries). C Distribution of different groups of genes (All (yellow; n = 21000 for mouse and 19288 for human; Housekeeping (HK; green; n = 3936 for mouse and 2779 for human); Developmental (Dev; purple; n = 962 for mouse and 1045 for human) within TADs according to Hi-C data previously generated in mESC13,19 and hESC118. Heatmap plot (with scaling by rows) showing the distribution of developmental (D) and housekeeping (E) genes within TADs previously identified in several human (n = 37 TAD maps) and mouse (n = 11 TAD maps) cell types.

(ii) Gene distribution: It was previously shown that TAD boundaries are enriched for housekeeping genes in comparison to tissue-specific genes66. However, from a regulatory standpoint, developmental genes (as defined here based on the presence of broad PcG domains around their TSS) and tissue-specific genes represent fundamentally different gene categories, as they typically differ in the type of promoter (i.e. CpG-rich for developmental genes and CpG-poor for tissue-specific genes) and long-range enhancer responsiveness65,67. To investigate the distribution of different types of genes (i.e. housekeeping, developmental, all) with respect to TADs, we divided TADs in 10 bins of equal sizes and calculated the number of genes within each bin based on the location of their TSS (Fig. 1B). In addition, genes located outside TADs (i.e. within TAD boundaries or inter-TAD genomic regions) were assigned to a bin labelled as bin 0 (Fig. 1B). Next, we computed the percentage of genes located in each bin. Overall, both housekeeping and developmental genes were preferentially enriched near TAD boundaries (i.e. bin 1), with housekeeping genes showing slightly higher percentage values for bin 1. In addition, housekeeping genes were often found not only close to but also within TAD boundaries (i.e. enriched in bin 1 and, depending on the cell type, also in bin 0), while developmental genes were preferentially located inside TADs and near their boundaries (i.e. enriched in bin 1 and depleted in bin 0) (Fig. 1C–E). Moreover, bin 1 housekeeping genes were located closer to TAD boundaries than their developmental counterparts (Fig. S4), which, considering the resolution of Hi-C data, is also in agreement with the more frequent location of housekeeping genes within TAD boundaries (i.e. bin 0). The preferential location of developmental genes close to TAD boundaries was similarly observed in both mice and humans, suggesting that it might represent an evolutionary conserved feature (Fig. 1C–E).

Sequential organization of developmental genes and clusters of CTCF sites at developmental boundaries

Based on the previous observations, we then evaluated whether the boundaries close to bin 1 developmental genes (i.e. developmental boundaries) display any distinctive features. First, we compared the number of CTCF peaks and CTCF ChIP-seq aggregated signals at developmental and non-developmental (i.e. other) TAD boundaries (developmental boundaries are defined as those having a developmental gene located in bin 1; Fig. 1B). For both CTCF metrics, the developmental boundaries showed higher values (Fig. 2A-B). Interestingly, despite these differences in CTCF binding, insulation scores and boundary strength (i.e. physical insulation) at developmental boundaries were similar to those observed at other TAD boundaries (Fig. 2A, B). Furthermore, similar analyses were performed by considering a broader set of developmental genes (i.e. genes included in the Gene Ontology (GO) term developmental process; GO:0032502), which were further divided in two groups depending on whether their promoter regions were covered or not by broad PcG domains (Fig. S5). These analyses showed that the insulation scores and boundary strength of the boundaries associated with developmental genes were similar regardless of whether their promoters were associated with broad PcG domains, suggesting that PcG domains do not have a major effect on the physical insulation properties of nearby TAD boundaries. Next, given previous reports indicating that paused RNA Pol2 at gene promoters can create boundaries21,22,25,68, we used PRO-seq data from mESC69 and GRO-seq data from hiPSC70 to calculate the RNA Pol2 pausing index (PI) for different gene categories (i.e. housekeeping, developmental, all) depending on their transcriptional status and proximity to TAD boundaries (i.e. bin1 genes) (Fig. S6). For each gene category, the PI were quite similar regardless of whether the genes were close (i.e. bin1 genes) or not to TAD boundaries (Fig. S6). Furthermore, in contrast to previous reports in Drosophila68,71, but in agreement with previous observations in mammalian cells72, developmental genes showed generally lower PI than either housekeeping or all genes regardless of their transcriptional status or proximity to TAD boundaries.

An external file that holds a picture, illustration, etc.
Object name is 41467_2024_51602_Fig2_HTML.jpg
Developmental genes and clusters of CTCF sites are sequentially organized near mouse TAD boundaries.

TAD maps, Hi-C data and CTCF ChIP-seq profiles previously generated in mESC (A) or hESC (B) were used to investigate the insulation scores, boundary strength, number of CTCF peaks and CTCF peaks aggregated signal at developmental TAD boundaries, all other TAD boundaries or random regions. Developmental boundaries were defined as those having a nearby developmental gene located within bin 1 (Fig. 1B). P-values were calculated using unpaired two-sided Wilcoxon tests with false discovery rate correction for multiple testing; Cliff’s delta (Cd) effect sizes are shown as coloured numbers (green: large effect size; blue: medium effect size; orange: small effect size; red: negligible effect size). In (A-B) box plots, the upper and lower parts of the box are the upper and lower quartiles, respectively, the horizontal line that splits the box in two is the median and the upper and lower whiskers indicate the maximum and minimum, respectively. C To investigate the distribution of CTCF peaks around different types of genes located close to TAD boundaries (Bin1 genes), we considered the TSS of each gene as a reference point and a window of ±100 Kb to calculate: ΔCTCFpeaks@Bdry as the difference between the number of CTCF peaks located towards the TAD center and those located towards the TAD boundary (negative values: CTCF peaks more abundant towards the TAD boundary; positive values: CTCF peaks more abundant towards the TAD center); ΔCTCFsignal@Bdry as the difference between the aggregated signal of the CTCF peaks located towards the TAD center and the aggregated signal of the CTCF peaks located towards the TAD boundary (negative values: CTCF signals higher towards the TAD boundary; positive values: CTCF signals higher towards the TAD center). D Histogram showing the distribution of ΔCTCFpeaks@Bdry values in mESC for different types of genes (developmental, housekeeping, all) located close to TAD boundaries (bin 1 in Fig. 1B). E Cumulative distribution plots for ΔCTCFpeaks@Bdry (left) and ΔCTCFsignal@Bdry (right) values in mESC. P-values were calculated using unpaired two-sided Wilcoxon tests with false discovery rate correction for multiple testing.

On the other hand, upon visual inspection of several developmental boundaries we noticed that developmental genes and CTCF clusters were sequentially organized (Figs. S12), with the developmental genes typically preceding most of the CTCF peaks (i.e. the CTCF peaks were preferentially located in the genomic regions separating the genes from the nearby TAD boundaries). To evaluate whether this sequential organization of developmental genes and CTCF clusters is somehow characteristic of developmental boundaries, we performed a global analysis of the distribution of CTCF peaks around genes located in bin 1 (Fig. 1B). Briefly, for each gene, we considered a window of +/−100 Kb around its TSS and compared the number of CTCF peaks (and associated CTCF ChIP-seq signals) located between the gene TSS and the nearby TAD boundary (i.e. outer window) with the number of peaks (and associated CTCF ChIP-seq signals) located between the gene TSS and the center of its TAD (i.e. inner window) (ΔCTCFpeaks@Bdry and ΔCTCFsignal@Bdry; Fig. 2C). Notably, the fraction of boundary-proximal genes in which the CTCF peaks and their associated signals were higher towards the TAD boundaries than towards the TAD centre (i.e. genes displaying negative ΔCTCFpeaks@Bdry and ΔCTCFsignal@Bdry values) was significantly larger for developmental genes than for other gene types (i.e. all, housekeeping) in both mice and humans (Fig. 2D, E; Fig. S7). This indicates that boundary-proximal genes in general and housekeeping genes in particular are frequently flanked by CTCF peaks, while for developmental genes the nearby CTCF peaks tend to accumulate in the genomic regions extending from the genes towards the TAD boundaries (i.e. developmental genes preceding the CTCF peaks). Next, using the same window of +/- 100 Kb around the TSS of bin 1 genes (i.e. inner and outer windows), we calculated the orientation of the CTCF sites relative to the TAD centers, distinguishing between CTCF sites oriented either towards (i.e. inward site) or away (i.e. outward site) from the TAD center (Fig. S8). A considerable fraction of CTCF peaks (at least 35%) was observed for both orientations regardless of the type of genes or windows analysed, supporting the potential relevance of not only inward but also outward CTCF sites for the proper establishment of intra-TAD chromatin interactions73. The differences in the fraction of inward and outward CTCF sites were negligible when comparing the inner and outer windows associated to developmental genes (Fig. S8). However, although still minor, non-negligible differences were observed for All and Housekeeping genes (Fig. S8), with the CTCF sites in the inner windows preferentially showing an inward orientation (~60%) and the CTCF sites in the outer window preferentially showing an outward orientation (>50%) (Fig. S8). Together with the results presented in Fig. 1D-E and Fig. S4, these results further suggest that housekeeping genes are often embedded within TAD boundaries where they are flanked by CTCF sites with divergent orientations74. In contrast, developmental genes tend to be located inside TADs but close to boundaries with large clusters of CTCF sites with complex motif orientations.

In summary, the sequential organization of developmental genes and CTCF clusters at developmental boundaries seems to represent a frequent and evolutionary conserved feature of mammalian genomes. To address the potential functional relevance of this sequential organization, we genetically dissected a couple of representative loci, Gbx2 and Six3/Six2 (Figs. S12, Fig. S9). These two loci were selected because they display the following features: (i) they contain developmental genes (i.e. Gbx2, Six3 and Six2) with broad PcG domains and large CGI clusters around their promoter regions; (ii) the developmental genes are located within gene-poor TADs and close to a TAD boundary (i.e. bin 1 genes); (iii) the developmental genes precede clusters of strong CTCF sites; (iv) the sequential organization of developmental genes and CTCF clusters is evolutionary conserved (Fig. S9).

The positioning of Gbx2 near a TAD boundary does not facilitate the maintenance of its own expression in ESC

We first focused on Gbx2, a major developmental gene with important functions in different processes such as neural patterning or naïve pluripotency75,76. Gbx2 is highly expressed in mESC (12.15 FPKM58), presumably due to the regulatory activity of a super-enhancer (SE) located approximately 60 Kb downstream of its TSS (Fig. 3A). Importantly, Gbx2 and its SE are found within a gene-poor TAD, with Gbx2 being located ~10 Kb upstream from three CTCF sites that constitute the 3’ TAD boundary (Fig. 3A). Moreover, this CTCF cluster separates Gbx2 from Asb18, a tissue specific gene located within the neighbouring TAD (~60 Kb downstream of the CTCF cluster) and that is inactive in mESC (0.056 FPKM58) (Fig. 3A). To start evaluating whether the sequential positioning of Gbx2 and the CTCF cluster at the 3’ boundary is of any functional relevance, we first generated multiple mESC clonal lines homozygous for the following re-arrangements: (i) Δ3xCTCF - a 10 Kb deletion that eliminates the three CTCF sites, which in principle should lead to the fusion of the Gbx2 and Asb18 TADs; (ii) 71Kb INV - a 71 kb inversion that re-positions Gbx2 and the SE with respect to the CTCF cluster, placing the enhancer in between Gbx2 and Asb18; (iii) Δ3xCTCF:71Kb INV - both the 71 Kb inversion and the 10 Kb deletion (Fig. 3B; Fig. S10). Next, we measured Gbx2 and Asb18 expression in all the generated mESC lines (Fig. 3C-D). Regarding Gbx2, we found that neither the 71Kb inversion nor the CTCF cluster deletion significantly affected its expression (Fig. 3C). The Gbx2 SE is located close to a CTCF site that is not included in the 71 Kb inversion (Fig. 3A, B) and that could theoretically contribute to the maintenance of proper Gbx2 expression levels in Δ3xCTCF and 71Kb INV cells. To directly address this possibility, we generated ESC lines with a small (256 bp) deletion that eliminates the CTCF site located upstream of the Gbx2 SE (ΔCTCF Gbx2SE) (Fig. S11A–C). The deletion of this CTCF site lead to a minor and non-significant decrease in Gbx2 expression levels (Fig. S11D). Overall, these results suggests that, in contrast to previous reports31,32,38, neither the positioning of Gbx2 close to the 3′ boundary nor the CTCF sites play a major role in the maintenance of Gbx2 expression.

An external file that holds a picture, illustration, etc.
Object name is 41467_2024_51602_Fig3_HTML.jpg
The positioning of Gbx2 close to its 3′ TAD boundary is not required to sustain its expression in mESC.

A mESC Hi-C data19 shows that Gbx2 and Asb18 are located within neighbouring TADs separated by a cluster of three CTCF sites122. The orientation of key CTCF sites is illustrated with red (sense) and blue (antisense) triangles. B Graphical overview of genomic rearrangements generated in mESC within the Gbx2/Asb18 locus: Δ3XCTCF; 71KbINV and Δ3XCTCF:71KbINV. C, D The expression of Gbx2 and Asb18 was measured by RT–qPCR in ESC that were either WT or homozygous for the genomic re-arrangements described in (B). Gbx2 and Asb18 expression was measured in the following number of biological replicates: WT −18 replicates; Δ3XCTCF −21 replicates using two different clonal lines; 71Kb INV −10 replicates using two different clonal lines; Δ3XCTCF:71Kb INV −14 replicates using two different clonal lines. P-values were calculated using two-sided unpaired t-tests (NS (not significant): fold-change < 2 or p > 0.05). E Capture-C experiments in WT, Δ3XCTCF, 71KbINV and Δ3XCTCF:71KbINV ESC using Gbx2 SE as a viewpoint (VP). Average Capture-C signals of the two replicates performed for each mESC line are shown around the Gbx2/Asb18 locus either individually (upper tracks) or after subtracting the WT Capture-C signals (lower tracks). The TAD boundary containing the three CTCF sites is highlighted in light blue. The red arrows indicate the 71 Kb inversion breakpoints. F The average Capture-C signals shown in (E) were measured within the Asb18 gene (chr1:89952677-90014577 (mm10)) and within five different 30 Kb control regions (Ctrls) located within the Gbx2 TAD (red asterisks in (E) indicate the midpoint of the controls regions (mm10): chr1:89803398-89833398; chr1:89753398-89783398; chr1:89703398-89733398; chr1:89653398-89683398; chr1:89603398-89633398). Capture-C signals are shown for the Δ3XCTCF, 71KbINV and Δ3XCTCF:71KbINV ESC as log2 fold-changes with respect to WT ESC. In (C, D) box plots, the upper and lower parts of the box are the upper and lower quartiles, respectively, the horizontal line that splits the box in two is the median and the upper and lower whiskers indicate the maximum and minimum, respectively. Source RT-qPCR data are provided as a Source Data file.

On the other hand, both the 71Kb inversion and the Δ3xCTCF deletion significantly increased Asb18 expression, with the inversion having a larger effect (Fig. 3D). Moreover, when both re-arrangements were combined, Asb18 expression levels were increased to a similar extent as with the inversion alone (Fig. 3D). Considering the established function of tandem CTCF sites as insulators77,78, the effects of the 10 Kb deletion on Asb18 expression were somehow expected. However, the results obtained for the inversion were more surprising and suggest that, when positioned close to the 3’ boundary, Gbx2 might display enhancer blocking activity and, thus, contribute to the physical insulation of its own regulatory domain. Interestingly, the Gbx2 gene is transcribed away from the boundary and towards the SE (Fig. 3A, B), which could potentially counteract cohesin-mediated loop extrusion2123 and, thus, contribute to enhancer blocking. To asses whether Gbx2 orientation contributes to the insulation of its own regulatory domain, we generated ESC lines in which we inverted the Gbx2 gene in the presence (Gbx2 INV) or absence (Gbx2 INV:Δ3xCTCF) of the 3XCTCF cluster (Fig. S12A–C). Analyses of the resulting ESC lines indicate that the orientation of Gbx2 does not significantly contribute to regulatory insulation, as the expression of Asb18 in Gbx2 INV and Gbx2 INV: Δ3xCTCF cells was rather similar to the one observed in WT and Δ3xCTCF cells, respectively (Fig. S12D). To more directly assess whether Gbx2 could contribute to physical insulation through enhancer blocking, we then performed Capture-C experiments using the Gbx2 SE or the Asb18 promoter as viewpoints in WT, Δ3xCTCF, 71Kb INV and Δ3xCTCF:71Kb INV ESC. As expected, the deletion of the 3XCTCF cluster reduced the physical insulation between the Gbx2 and Asb18 TADs, thus increasing the contact frequency between Asb18 and the SE (Fig. 3E, F; Fig. S13C, D) as well as between Asb18 and Gbx2 (Fig. S13C, E). In contrast, the 71 Kb inversion strongly increased the contact frequency between the SE and the 3XCTCF cluster (Fig. 3E, Fig. S13B), but had a minor impact on the Asb18-SE or Asb18-Gbx2 contacts (Fig. 3E, F, Fig. S13C–E). Furthermore, both the 3XCTCF deletion and the 71 Kb inversion reduced the contact frequency between the SE and Gbx2 (Fig. 3E, Fig. S13A), which, nevertheless, did not have major regulatory effects on Gbx2 expression (Fig. 3C). Overall, these Capture-C experiments suggest that the increased expression of Asb18 in cells with the 71 Kb inversion is unlikely to be caused by the loss of Gbx2 enhancer blocking activity. One alternative explanation is that the inversion reduces the linear distance between Asb18 and the SE (from 141 Kb to 83 Kb), which in turn might increase Asb18 expression without significantly affecting enhancer-gene contacts7981. In agreement with this non-linear relationship between gene activity and E-P contact frequency, the combination of the 71 Kb inversion and the CTCF deletion resulted in a strong loss of physical insulation between the Gbx2 and Asb18 TADs that was not translated into a further increase in Asb18 expression compared to cells with the inversion alone (Fig. 3D–F, Fig. S13). Furthermore, since Asb18 is expressed at low levels in mESC (0.056 FPKM), the upregulation of Asb18 in absolute levels in [increment]3XCTCF (~5-fold) or 71Kb INV (~16-fold) cells is still very small, suggesting that the responsiveness between the Asb18 promoter and the Gbx2 enhancer might be limited67.

Robust regulatory insulation between the Gbx2 and Asb18 TADs depends on the cooperative effects of Gbx2 and nearby CTCF sites

The previous results indicate that the physical insulation between the Gbx2 and Asb18 TADs can be largely attributed to the cluster of CTCF sites. Interestingly, despite increasing Asb18-SE contact frequency, the combination of the CTCF deletion and the 71 Kb inversion did not significantly change Asb18 expression compared to the cells with the 71 Kb inversion alone (Fig. 3D–F; Fig. S13). Since promoter competition can occur regardless of the relative position of the shared enhancer(s) with respect to the competing promoters47,48, this made us wonder whether, in the absence of the CTCF sites, the presence of Gbx2 could prevent a further increase in Asb18 expression through promoter competition rather than enhancer blocking47,48. To test this hypothesis, we generated mESC lines with the following homozygous re-arrangements (Fig. S14): (i) ΔPromGbx2 - a small (1 Kb) deletion spanning the Gbx2 promoter that reduces Gbx2 expression (Fig. 4A, B) and (ii) Δ3XCTCF:ΔPromGbx2 - deletions spanning the Gbx2 promoter and the CTCF cluster, respectively (Fig. 4A). Importantly, the 1 Kb promoter deletion should theoretically disrupt both the enhancer-blocking and promoter competition capacity of Gbx2, while barely changing the linear distance between Asb18 and the SE (Fig. 4A). The promoter deletion alone resulted in elevated Asb18 expression levels (Fig. 4C). Most interestingly, the combination of the Gbx2 promoter and CTCF cluster deletions lead to a strong and synergistic increase in Asb18 expression (61.8 fold-change in Δ3xCTCF:ΔPromGbx2 vs WT; 5.5 and 7.5 fold-changes in Δ3xCTCF and ΔPromGbx2 vs WT, respectively) (Fig. 4C), resulting in Asb18 expression levels that were considerably higher than those observed in cells with both the CTCF deletion and the 71 Kb inversion (61.8 fold-change in Δ3xCTCF:ΔPromGbx2 vs WT; 17.6 fold-change in Δ3xCTCF:71 Kb INV vs WT) (Fig. 3Dvs Fig. 4C). Next, we also performed Capture-C experiments in these cell lines using again the Gbx2 SE and the Asb18 promoters as viewpoints. Remarkably, the deletion of the Gbx2 promoter, either alone or in combination with the CTCF cluster deletion, showed minor effects on Asb18-SE contact frequency in comparison to WT and Δ3xCTCF cells, respectively (Fig. 4D, E; Fig. S15B, C). Furthermore, the deletion of the Gbx2 promoter did not have major effects on the interaction frequency between either Gbx2 or Asb18 and the 3XCTCF cluster (Fig. 4D, Fig. S15A, B, D). Accordingly, RAD21 ChIP-seq experiments showed that cohesin profiles within the Asb18 locus were not significantly affected by the Gbx2 promoter deletion (Fig. S16A). On the other hand, H3K27ac ChIP-seq signals at the Gbx2 SE were rather similar among all the generated cell lines, albeit slightly elevated in the CTCF deletion cells (Fig. S16B), arguing that the observed Asb18 expression changes are not caused by differences in enhancer activity.

An external file that holds a picture, illustration, etc.
Object name is 41467_2024_51602_Fig4_HTML.jpg
Gbx2 and the nearby CTCF cluster cooperatively contribute to the regulatory insulator capacity of the nearby TAD boundary.

A Graphical overview of different genomic rearrangements generated in mESC within the Gbx2/Asb18 locus: Δ3XCTCF (same as in Fig. 3B); ΔPromGbx2; Δ3XCTCF:ΔPromGbx2. The expression of Gbx2 (B) and Asb18 (C) was measured by RT–qPCR in ESC that were either WT or homozygous for the genomic re-arrangements described in (A). For each cell line, Gbx2 and Asb18 expression was measured in the following number of biological replicates: WT −18 replicates (same as in Fig. 3C, D); Δ3XCTCF −21 replicates using two different clonal lines (same as in Fig. 3C, D); ΔPromGbx2 -5 replicates for Gbx2 and 6 replicates for Asb18 using two different clonal lines; Δ3XCTCF:ΔPromGbx2 -6 replicates using two different clonal lines. Expression differences among ESC lines were calculated using two-sided unpaired t-tests (NS (not significant): fold-change < 2 or p > 0.05). D Capture-C experiments in WT, Δ3XCTCF, ΔPromGbx2 and Δ3XCTCF:ΔPromGbx2 ESC using the Gbx2 SE as a viewpoint. The average Capture-C signals of the two replicates performed for each mESC line are shown around the Gbx2/Asb18 locus individually (upper tracks) or after subtracting the WT signals (lower tracks). The three CTCF sites deleted within the Gbx2/Asb18 TAD boundary are highlighted in light blue. Capture-C tracks for WT and Δ3XCTCF ESC are the same as in Fig. 3E. E The average Capture-C signals shown in (D) were measured within the Asb18 gene (highlighted in yellow in (D); chr1:89952677-90014577 (mm10) as well as within five different 30 Kb control regions (Ctrls) located within the Gbx2 TAD (red asterisks in (D) indicate the midpoint of the same control regions as in Fig. 3F). Capture-C signals are shown for the Δ3XCTCF, ΔPromGbx2 and Δ3XCTCF:ΔPromGbx2 ESC as log2 fold-changes with respect to WT ESC. In (B, C) box plots, the upper and lower parts of the box are the upper and lower quartiles, respectively, the horizontal line that splits the box in two is the median and the upper and lower whiskers indicate the maximum and minimum, respectively. Source RT-qPCR data are provided as a Source Data file.

Overall, these results support the notion of Gbx2 preferentially contributing to regulatory (rather than physical) insulation through promoter competition. Importantly, our data also suggest that Gbx2 and the CTCF cluster cooperatively confer the nearby 3’ TAD boundary with strong insulator capacity (Table 1).

Table 1

Summary of the main results obtained for the Gbx2/Asb18 locus

Cell line nameCell line descriptionGene expression analysesCapture-C experimentsConclusions
Δ3xCTCF (Fig. (Fig.3;3; Fig. S13)10 Kb deletion that eliminates the three CTCF sites separating the Gbx2 and Asb18 TADs.

Gbx2 expression is not affected.

Asb18 expression is significantly increased (~5-fold).

The contact frequency between the Gbx2 SE and Gbx2 is reduced.

The contact frequency between the Gbx2 SE and Asb18 is increased.

The positioning of Gbx2 close to the 3XCTCF cluster does not play a major role in the maintenance of Gbx2 expression in ESC.

The CTCF cluster strengthens the physical insulation of the Gbx2 regulatory domain, preventing spurious enhancer-gene contacts.

71Kb INV (Fig. (Fig.3;3; Fig. S13)Inversion that re-positions Gbx2 and its SE with respect to the CTCF cluster, placing the SE in between Gbx2 and Asb18.

Gbx2 expression is not affected.

Asb18 expression is significantly increased (~16-fold).

The contact frequency between the Gbx2 SE and Gbx2 is reduced.

Minor impact on Asb18-Gbx2 SE contacts.

Strong increase in the contact frequency between the Gbx2 SE and the 3XCTCF cluster.

The positioning of Gbx2 close to the 3xCTCF cluster does not play a major role in the maintenance of Gbx2 expression in ESC.

Gbx2 does not contribute to physical insulation through enhancer blocking. The increased expression of Asb18 might be explained by the reduced linear distance between Asb18 and the Gbx2 SE in 71Kb INV cells.

Δ3xCTCF:71Kb INV (Fig. (Fig.3;3; Fig. S13)Cells with both the Δ3xCTCF deletion and the 71 Kb inversion.

Gbx2 expression is not affected.

Asb18 expression is increased to similar levels as in 71Kb INV cells.

The contact frequency between the SE and Gbx2 is reduced.

The contact frequency between the Gbx2 SE and Asb18 is strongly increased.

Non-linear relationship between gene activity and E-P contact frequency: the increase in Gbx2 SE-Asb18 contacts does not lead to a further increase in Asb18 expression.
ΔProm Gbx2 (Fig. (Fig.4;4; Fig. S15)1 Kb deletion spanning the Gbx2 promoter.Asb18 expression is significantly increased (~7-fold).Minor impact on Asb18-Gbx2 SE contacts.Gbx2 contributes to regulatory rather than physical insulation through promoter competition.
Δ3XCTCF:ΔProm Gbx2 (Fig. (Fig.4;4; Fig.  S15)Cells with both the Δ3xCTCF and ΔPromGbx2 deletions.Strong and synergistic increase in Asb18 expression (~60-fold).The contact frequency between the Gbx2 SE and Asb18 is increased to a similar extent as in Δ3xCTCF cells.Gbx2 contributes to regulatory rather than physical insulation through promoter competition. Gbx2 and the CTCF cluster cooperatively confer the nearby TAD boundary with strong insulator capacity.

A large CTCF cluster prevents promoter competition between Six3 and Six2

In order to test whether the previous observations could be extended to other developmental boundaries and cellular contexts, we then generated a similar set of genetic re-arrangements within the Six3/Six2 locus (Fig. 5A). Six3 and Six2 are two typical developmental genes (i.e. with broad PcG domains and large CGI clusters around their promoters) that are located close to each other in linear space (~68 Kb between Six3 and Six2). However, there is a strong and conserved TAD boundary containing seven CTCF sites and spanning ~50 Kb that separates the Six3 and Six2 regulatory domains (Fig. 5A)82,83. Accordingly, Six3 and Six2 display largely non-overlapping expression patterns during embryogenesis: Six3 is expressed in the developing forebrain or the eye, while Six2 shows high expression in the developing kidney or the facial mesenchyme82,83. We previously showed that within the Six3 TAD there is a conserved SE that specifically activates Six3, but not Six2, in neural progenitors (NPC) and the developing forebrain (Six3 expression in NPC: 10.33 FPKM; Six2 expression in NPC: 0.21 FPKM)58,63. With this information at hand, we first generated mESC lines with the following homozygous re-arrangements: (i) 156Kb INV - 156 Kb inversion between Six3 and its SE that places the enhancer close to the TAD boundary and in between Six3 and Six2; (ii) Δ6XCTCF - 36 Kb deletion that eliminates six out of the seven CTCF sites separating Six3 and Six263; (ii) Δ6XCTCF:156Kb INV - both the 156 Kb inversion and the 36 Kb deletion (Fig. 5B; Fig. S17). Once multiple clonal mESC lines were obtained for each of these re-arrangements, we differentiated them into NPC and measured the expression of Six3 and Six2 (Fig. 5C, D).

An external file that holds a picture, illustration, etc.
Object name is 41467_2024_51602_Fig5_HTML.jpg
The large CTCF cluster separating the Six3 and Six2 TADs prevents promoter competition between these two genes.

A NPC Hi-C19 data shows that Six3 and Six2 are located in neighbouring TADs separated by a cluster of seven CTCF sites122. The orientation of key CTCF sites is illustrated with red (sense) and blue (antisense) triangles. B Graphical overview of genomic rearrangements generated in mESC within the Six3/Six2 locus: Δ6XCTCF; 156KbINV; Δ6XCTCF:156KbINV. C, D The expression of Six3 and Six2 was measured by RT–qPCR in NPC that were WT or homozygous for the genomic re-arrangements described in (B). For each cell line, Six3 and Six2 expression was measured in the following number of biological replicates: WT -16 replicates; Δ6XCTCF -19 replicates using a previously characterized clonal line63; 156KbINV −10 replicates for Six3 and 14 replicates for Six2 using two different clonal lines; Δ6XCTCF:156KbINV −18 replicates using three different clonal lines. E Graphical overview of the 226KbINV mESC line. F Six3 and Six2 expression was measured by RT–qPCR in NPC that were WT or homozygous for the 226KbINV. For each cell line, Six3 and Six2 expression was measured in the following number of biological replicates: WT -4 replicates; 226KbINV -4 replicates using two different clonal lines. G Graphical overview of the Δ6XCTCF:Six2−/− mESC line. H Six3 expression was measured by RT–qPCR in NPC that were either WT or homozygous for the Δ6XCTCF and Δ6XCTCF:Six2−/− deletions. For each cell line, Six3 was measured in the following number of biological replicates: WT −16 replicates (same as in C); Δ6XCTCF −19 replicates (same as in C); Δ6XCTCF:Six2−/− -11 replicates using three different clonal lines. Expression differences among cell lines were calculated using two-sided unpaired t-tests (NS (not significant): fold-change < 2 or p > 0.05). In (C, D, F, H) box plots, the upper and lower parts of the box are the upper and lower quartiles, respectively, the horizontal line that splits the box in two is the median and the upper and lower whiskers indicate the maximum and minimum, respectively. Source RT-qPCR data are provided as a Source Data file.

Similarly to our results for the Gbx2 locus, the 156 Kb inversion did not significantly affect Six3 expression (Fig. 5C). It is worth noting that this inversion places Six3 close to a single CTCF peak located next to the SE (Fig. 5A). Although we previously showed that this CTCF site was not required for Six3 induction in NPC63, it could theoretically contribute to Six3 expression in cells with the inversion. Therefore, we engineered a 226 Kb inversion that places Six3 further away from the TAD boundary while preserving the linear distance with respect to the enhancer (Fig. 5E; Fig. S18). Notably, this 226 Kb inversion did not affect Six3 induction in NPC either (Fig. 5F). These results further suggest that the location of developmental genes close to TAD boundaries/CTCF clusters does not universally facilitate their own expression. Furthermore, and in contrast to our findings for the Gbx2 locus, neither the 156 Kb nor the 226 Kb inversion affected Six2, which remained lowly expressed in NPC (Fig. 5D, F). This could be explained by the differences between the CTCF clusters present at each locus (seven CTCF sites spanning ~50 Kb at the Six3/Six2 locus vs three CTCF sites spanning ~10 Kb at the Gbx2/Asb18 locus), which is significantly larger in the Six3/Six2 locus and, thus, could confer stronger physical insulation.

On the other hand, and in contrast to our observations for the Gbx2/Asb18 locus, the deletion of the CTCF cluster not only led to the ectopic activation of Six2 (Fig. 5D), but also significantly impaired Six3 induction in NPC (Fig. 5C). Considering the results obtained with the inversions described above, the reduced expression of Six3 can not be simply attributed to a potential role of the CTCF sites as facilitators of enhancer-gene communication. Recent work in Drosophila indicates that insulators protect genes from spurious interactions with not only enhancers but also silencers located within neighbouring TADs84. Therefore, the CTCF cluster might protect Six3 from the repressive effects of putative silencer elements located within the Six2 TAD and/or from the promoter competition activity of Six2. Since there are no robust chromatin signatures to globally identify silencers, we decided to test the Six2 promoter competition hypothesis by generating mESC lines with deletions spanning both the CTCF cluster and Six2 (Fig. 5G; Fig. S19). Interestingly, the Six2 deletion rescued, albeit partly, the impaired induction of Six3 in NPC (Fig. 5H). This suggests that, rather than facilitating the communication between Six3 and its cognate enhancers, the CTCF cluster protects Six3 from promoter competition by Six2. However, since the Six2 deletion led to a partial rather than total rescue of Six3 expression levels, the CTCF cluster might also protect Six3 from putative silencers found within the Six2 TAD84.

Cooperative insulation of the Six3 and Six2 regulatory domains by the CTCF cluster and promoter competition

The previous results suggest that, in the absence of the CTCF cluster, Six2 competes with Six3 for the NPC enhancers located within the Six3 TAD. Therefore, we wondered whether this promoter competition could be reciprocal and, thus, Six3 could participate, together with the CTCF cluster, in the robust insulation of the Six3 regulatory domain. To explore this possibility, we generated mESC lines with (i) a 27 Kb deletion spanning the Six3 gene (i.e. Six3−/−); (ii) deletions spanning both Six3 and the CTCF cluster (i.e. Δ6XCTCF:Six3−/−) (Fig. 6A; Fig. S20). All the resulting clonal ESC lines were differentiated into NPC, in which we measured Six2 and Six3 expression (Fig. 6A-B).

An external file that holds a picture, illustration, etc.
Object name is 41467_2024_51602_Fig6_HTML.jpg
Six3 contributes to the robust insulation of the Six3 and Six2 regulatory domains through promoter competition.

A Genomic rearrangements generated in mESC within the Six3/Six2 locus: Δ6XCTCF; Six3−/−; Δ6XCTCF: Six3−/−; Δ4XCTCF; Δ4XCTCF:Six3−/−. B, C Six3 and Six2 expression was measured by RT–qPCR in NPC homozygous for the indicated re-arrangements using the following number of biological replicates: WT −16 replicates (same as in Fig. 5C, D); Δ6XCTCF −19 replicates using a previously characterized clonal line63 (same as in Fig. 5C, D); Six3−/− −16 replicates using three clonal lines; Δ6XCTCF:Six3−/− -15 replicates using three clonal lines. D Capture-C experiments in WT, Δ6XCTCF, Six3−/− and Δ6XCTCF:Six3−/− NPC using Six3 SE as a viewpoint. Average Capture-C signals of the two replicates performed for each cell line are shown around the Six3/Six2 locus individually (upper tracks) or after subtracting the WT signals (lower tracks). E Average Capture-C signals were measured around Six2 (highlighted in yellow in (D); chr17:85674267-85698254 (mm10)) and within five 30 Kb control regions (Ctrls) located within the Six3 TAD (red asterisks in (D) indicate the midpoint of the controls regions (mm10): chr17:85388601-85418601; chr17:85338601-85368601, chr17:85288601-85318601, chr17:85238601-85268601, chr17:85188601-85218601). Capture-C signals are shown for the Δ6XCTCF, Six3−/− and Δ6XCTCF:Six3−/− NPC as log2 fold-changes with respect to WT NPC. F, G Six3 and Six2 expression was measured by RT–qPCR in NPC homozygous for the indicated genomic re-arrangements using the following number of biological replicates: WT −16 replicates (same as in Fig. 5C, D); Six3−/− −16 replicates using three different clonal lines (same as in Fig. 6C); Δ4XCTCF −12 replicates for Six3 and 11 replicates for Six2 using three different clonal lines; Δ4XCTCF:Six3−/− −12 replicates using three different clonal lines. Expression differences among cell lines were calculated using two-sided non-paired t-tests (NS (not significant): fold-change < 2 or p > 0.05); ND (not detectable)). In (B, C, F, G) box plots, the upper and lower parts of the box are the upper and lower quartiles, respectively, the horizontal line that splits the box in two is the median and the upper and lower whiskers indicate the maximum and minimum, respectively. Source RT-qPCR data are provided as a Source Data file.

Remarkably, although the Six3 deletion alone mildly changed (albeit in a non-statistically significant manner) Six2 expression in NPC, Six2 expression was strongly and synergistically increased when the Six3 and CTCF cluster deletions were combined (Fig. 6C) (72.1 Fold-change in Δ6xCTCF:Six3−/− vs WT; 8.8 fold change in Δ6xCTCF and 1.7 fold-change in Six3−/− vs WT, respectively), thus in agreement with the contribution of Six3 to regulatory insulation. Next, we performed Capture-C experiments in these cell lines using the Six3 SE or the Six2 promoter as viewpoints. As expected, the deletion of the CTCF cluster led to a loss of physical insulation between the Six3 and Six2 TADs and increased the contact frequency between Six2 and the SE (Fig. 6D, E; Fig. S21C, D). In addition, this deletion strongly increased the contacts between Six3 and Six2 (Fig. S21C, E), in agreement with the formation of a shared transcriptional hub51. In contrast, the CTCF cluster deletion did not affect the contact frequency between the SE and Six3 (Fig. 6D, Fig. S21A), further indicating that the 6XCTCF cluster does not play a major role as a facilitator of Six3-SE communication in NPC. Notably, the deletion of Six3 did not change the Six2-SE contact frequency in comparison to WT cells (Fig. 6D, E; Fig. S21C, D) and only led to a minor increase in the interaction frequency between either the SE or Six2 and the 6XCTCF cluster (Fig. 6D, Fig. S21B, C, F). Finally, the combined deletion of Six3 and the CTCF cluster, which resulted in a strong induction of Six2, mildly increased the Six2-SE contact frequency in comparison to the CTCF deletion alone (Fig. 6D, E; Fig. S21C, D). Altogether, these results suggest that, as observed for the Gbx2/Asb18 locus, Six3 preferentially contributes to regulatory insulation through promoter competition, while Six3-dependent enhancer blocking seems to have a comparably smaller, albeit non-negligible, contribution. Therefore, enhancer blocking (i.e. physical insulation) is mostly dependent on the CTCF cluster, which together with Six3-mediated promoter competition cooperatively confer the TAD boundary with strong insulator capacity. Importantly, the contribution of Six3 to regulatory insulation through promoter competition can also explain why the 156Kb and 226Kb inversions described above did not have any major impact on Six2 expression either alone or in combination with the CTCF cluster deletion (Fig. 5D, F). In agreement with this model, RAD21 ChIP-seq experiments showed that the CTCF cluster deletion increased cohesin levels around Six2, particularly at a CTCF site located upstream of this gene (red asterisk in Fig. S22A), while cohesin profiles were not affected by the Six3 deletion (Fig. S22A). On the other hand, H3K27ac ChIP-seq signals at the Six3 SE were rather similar among all the generated cell lines, except in the Six3−/− cells in which we observed higher H3K27ac levels, arguing that the observed Six2 expression changes are not caused by differences in enhancer activity (Fig. S22B).

Overall, our data suggest that, as for the Gbx2/Asb18 locus, Six3 and the CTCF cluster cooperatively contribute to the robust insulation of the Six3 and Six2 regulatory domains (Table 2).

Table 2

Summary of the main results obtained for the Six3/Six2 locus

Cell line nameCell line descriptionGene expression analysesCapture-C experimentsConclusions
Δ6xCTCF (Figs. (Figs.55 and and6;6; Fig. S21)36 Kb deletion that eliminates six out of the seven CTCF sites separating the Six3 and Six2 TADs.

Six3 induction is significantly impaired (~5-fold).

Six2 expression is significantly increased (~9-fold).

The contact frequency between the Six3 SE and Six3 is not affected.

Increased contact frequency between Six3 and Six2.

The contact frequency between the Six3 SE and Six2 is increased.

The positioning of Six3 close to the CTCF cluster might facilitate Six3 induction, although changes in Six3-SE contact frequency are not observed.

The CTCF cluster strengthens the physical insulation of the Six3 regulatory domain, preventing spurious enhancer-gene contacts.

156KbINV (Fig. (Fig.55)Inversion that re-positions Six3 and its SE with respect to the CTCF cluster, placing the SE in between Six3 and Six2.

Six3 expression is not affected.

Six2 expression is not affected.

NA

The positioning of Six3 close to the CTCF cluster does not facilitate Six3 induction. The impaired induction of Six3 in Δ6xCTCF might involve alternative mechanisms.

Six3 does not contribute to the insulation of its regulatory domain through enhancer blocking.

Δ6xCTCF: 156KbINV (Fig. (Fig.55)Cells with both the Δ6xCTCF deletion and the 156 Kb inversion.

Six3 induction is significantly impaired.

Six2 expression is increased to similar levels as in Δ6xCTCF.

NASix3 does not contribute to the insulation of its regulatory domain through enhancer blocking.
Δ6xCTCF:Six2−/−(Fig. (Fig.55)Cells with both the Δ6xCTCF deletion and a 11 Kb deletion spanning the Six2 gene.Six3 induction is partially rescued (~2-fold reduction) in comparison to Δ6xCTCF cells (~5-fold reduction).NAThe CTCF cluster might protect Six3 from promoter competition by Six2 and, potentially, from putative silencers found within the Six2 TAD (speculative).
Six3−/− (Fig. (Fig.6;6; Fig. S21)27 Kb deletion spanning the Six3 gene.Six2 expression is not affected.Minor impact on Six2-Six3 SE contacts.Six3 does not contributes to the physical insulation of its regulatory domain.
Δ6XCTCF:Six3−/− (Fig. (Fig.6;6; Fig. S21)Cells with both the Δ6xCTCF deletion and the Six3−/− deletion.Strong and synergistic increase in Six2 expression (~70-fold).The contact frequency between the Six3 SE and Six2 is mildly increased in comparison to Δ6xCTCF cells.

Six3 contributes to regulatory rather than physical insulation through promoter competition.

Six3 and the CTCF cluster cooperatively confer the nearby TAD boundary with strong insulator capacity.

Δ4xCTCF (Fig. (Fig.66)36 Kb deletion that eliminates four out of the seven CTCF sites separating the Six3 and Six2 TADs.

Six3 induction is significantly impaired.

Six2 expression is not affected.

NAThe CTCF cluster might protect Six3 from promoter competition by Six2 and, potentially, from putative silencers found within the Six2 TAD (speculative).
Δ4XCTCF:Six3−/− (Fig. (Fig.66)Cells with both the Δ4xCTCF and Six3−/− deletions.Strong and synergistic increase in Six2 expression (~16-fold).NASix3 and the CTCF cluster cooperatively confer the nearby TAD boundary with strong insulator capacity.

Large CTCF clusters prevent competition between adjacent developmental genes

One major difference between the results obtained for the Gbx2/Asb18 and Six3/Six2 loci is that the deletion of the corresponding CTCF clusters impaired the induction of Six3, but did not significantly affect the expression of Gbx2. We hypothesized that this could be attributed to the fact that Six2 and Six3 are both typical developmental genes, which are characterized by having promoters with large clusters of CpG islands and strong long-range enhancer responsiveness (Fig. 5A)63,65,67. On the contrary, while Gbx2 is also a developmental gene with a large CGI cluster around its promoter, Asb18 is a tissue specific gene with a CpG-poor promoter, which typically show weak long-range enhancer responsiveness (Fig. 3A)63,67. Consequently, in the absence of CTCF sites, Six2 and Six3 might engage into strong promoter competition for shared enhancers, while Asb18 might represent a weak competitor against Gbx2. Furthermore, the regulatory domains of developmental genes, such as Six2, might often contain silencers whose long-range repressive capacity needs to be properly insulated84,85. Therefore, we hypothesize that the boundary insulating the Six3 and Six2 regulatory domains might require a particularly large CTCF cluster (seven CTCF sites spanning ~50 Kb) in comparison to the one found between Gbx2 and Asb18 (three CTCF sites spanning ~10 Kb) (Figs. 3A and and5A).5A). To test this prediction, we generated mESC lines with a deletion that removed four out of the seven CTCF sites separating the Six3 and Six2 domains (Fig. 6A; Fig. S23). In addition, we also generated mESC lines with the partial deletion of the CTCF cluster plus the Six3 deletion (Fig. 6A). Interestingly, the partial deletion of the CTCF cluster alone did not significantly increase Six2 expression, but strongly impaired Six3 induction (Fig. 6F, G). Together with the results obtained for the inversions and the 6XCTCF deletion described above (Fig. 5), this further suggests that, rather than facilitating Six3 expression, the CTCF cluster separating the Six3 and Six2 TADs might protect Six3 from promoter competition by Six2 and, potentially, from the repressive effects of putative silencers located within the Six2 TAD33,84, although the latter possibility remains speculative and would require further experimental evidences. Last but not least, the combination of the partial deletion of the CTCF cluster and the Six3 deletion led to a strong and synergistic induction of Six2 in NPC (Fig. 6G). This further supports that Six3 contributes to regulatory insulation through promoter competition and, more generally, that the robust insulation provided by developmental boundaries requires the cooperative contribution of CTCF clusters and transcriptionally active developmental genes (Tables 12).

Discussion

During embryogenesis, developmental genes are typically expressed in several spatial and/or temporal contexts, which is enabled by the presence of multiple, distinct and highly specific enhancers within regulatory domains delimited by insulators (e.g. CTCF sites)3,86. Moreover, within a particular cellular context, the expression of developmental genes is often controlled by enhancer clusters (i.e. SE, locus control regions (LCR)) with strong regulatory activity87. However, CTCF sites might not be sufficient to restrain the regulatory activity of these complex enhancer landscapes towards their target genes33,34, suggesting that additional mechanisms might be necessary to ensure gene expression specificity and the robust insulation of developmental TADs. Here we report that developmental TADs are gene-poor and often delimited by rather unique boundaries. More specifically, we show that, within these TADs, developmental genes and CTCF clusters are often sequentially organized (with CTCF peaks being predominantly located between genes and TAD boundaries) and cooperatively strengthen the regulatory insulation capacity of nearby boundaries, thus contributing to the establishment of specific expression patterns during development.

The large size and low gene density of developmental TADs might be required to accommodate the complex enhancer landscapes responsible for the specific, yet diverse, spatiotemporal expression patterns that many developmental genes display during embryogenesis3,86,88. Furthermore, due to the long-range responsiveness of developmental gene promoters63,67, their cognate enhancers can be separated by large linear distances and, thus, located anywhere within developmental TADs. In contrast, the regulatory elements controlling the expression of either housekeeping or tissue-specific genes tend to be proximal or even embedded within promoter regions89,90. Moreover, as housekeeping and tissue-specific genes show lower long-range enhancer responsiveness than developmental genes, they might be able to co-exist with other genes with similar promoter types within gene-rich TADs without interfering with each other’s expression profiles14,91.

Here we show that developmental genes and CTCF clusters cooperatively strengthen the insulation capacity of nearby boundaries (i.e. developmental boundaries). In agreement with previous work, the physical insulation of the investigated TADs was mostly dependent on the CTCF clusters36,77,78,92. In contrast, the investigated developmental genes do not seem to act as strong physical barriers preventing ectopic enhancer-gene communication (i.e. enhancer blocking), but preferentially contribute to regulatory insulation through promoter competition. The contribution of developmental genes to regulatory insulation through non-structural mechanisms is also in agreement with the fact that developmental boundaries show similar insulation scores (i.e. physical insulation) to those observed for TAD boundaries in general (Fig. 2A, B). For developmental genes located close to TAD boundaries, the majority of their cognate enhancers should be located at more central positions within the same TAD. Consequently, the linear distance between the boundary-proximal developmental genes and their cognate enhancers should be often smaller than the distance between those same enhancers and non-target genes located at the other side of the boundary within the adjacent TAD. Considering that short linear distances facilitate the functional communication between genes and enhancers41,80,81, we hypothesize that the shorter linear distances separating boundary-proximal genes and their cognate enhancers might give those developmental genes a competitive advantage over non-target genes located on the other side of the developmental boundary93. Recent studies indicate that the physical insulation provided by CTCF boundaries is dynamic and partial, with strong enhancers, such as those typically controlling the expression of major developmental genes, being able to bypass those boundaries and activate genes across TADs3336. At the single cell level, the permeability of CTCF boundaries might result in a small fraction of cells/alleles in which cohesin-mediated loops bypass those boundaries and enable spurious contacts between enhancers and non-target genes. In the context of developmental loci, such as those investigated in our work, this can lead to the formation of multiway contacts (hubs) in which enhancers can simultaneously interact with their target developmental genes as well as with non-target genes located in neighboring domains51. However, we speculate that the strong enhancer responsiveness of developmental genes63,64 together with shorter linear distances with respect to their cognate enhancers93 might give developmental genes a competitive advantage over non-target genes and, thus, prevent the spurious contacts between enhancers and non-target genes from being productive.

At the molecular level, it is currently unclear which mechanisms might explain how active genes contribute to insulation through promoter competition. Recent work suggests that RNA Pol2 can act as a weak physical barrier against cohesin-mediated loop extrusion2123,25, although the overall contribution of RNA Pol2 to chromatin architecture and physical insulation remains controversial28,29. In this regard, forced activation of candidate genes in ESC does not cause changes in chromatin insulation, suggesting that transcription and RNA Pol2 recruitment are not sufficient for the emergence of TAD boundaries19. Moreover, the potential role of RNA Pol2 as a loop extrusion barrier can not explain how developmental genes contribute to insulation through promoter competition, as this does not require the placement of developmental genes in between enhancers and non-target genes (Figs. 3 and and55)47,48. Instead, promoter competition might involve non-structural mechanisms whereby multiple promoters compete for rate-limiting factors (e.g. TFs, GTFs, RNA Pol2) present within shared transcriptional hubs56,57,94. Although these models and the putative rate-limiting factors await experimental validation, we speculate that nearby promoters with strong enhancer responsiveness and similar architecture (e.g. Six3 and Six2) might be particularly sensitive to promoter competition unless insulated by large CTCF clusters. On the other hand, the positioning of developmental genes close to the boundaries should often place them in between their cognate enhancers and non-target genes located in the adjacent domain, thus potentially enabling developmental genes to display enhancer blocking function. In this regard, it is possible that the developmental genes investigated in this study (i.e. Gbx2 and Six3) act as weak enhancer blockers whose effects on physical insulation can not be easily detected due to the current resolution and sensitivity of Capture-C and other 3C methods, but that, nevertheless, can still result in transcriptional changes80,95. Furthermore, in the context of boundaries close to strongly expressed genes (e.g. globin genes), the contribution of active transcription and RNA Pol2 complexes to physical insulation might be larger24,4346 in comparison to most developmental genes that, even when active, are expressed at comparably moderate levels.

It has been previously proposed that TAD boundaries and the associated CTCF clusters can (i) facilitate the communication between genes and enhancers located within the same domain and (ii) insulate genes from the regulatory activity of enhancers located in other domains. Although CTCF clusters might facilitate ultra long-range enhancer-gene contacts (e.g. Shh, Sox9)39,96, at shorter distances (such as those separating Gbx2 and Six3 from their SEs in ESC and NPC, respectively) CTCF sites seem to be dispensable and other types of tethering elements (e.g. CpG islands in vertebrates, GAGA elements in Drosophila) can mediate intra-TAD enhancer-gene communication63,84. However, although proper expression of Gbx2 and Six3 in ESC and NPC, respectively, does not seem to require the proximity of these genes to CTCF clusters, it is certainly possible that the CTCF sites facilitate the communication of Gbx2 and/or Six3 with more distal enhancers in other cellular contexts. Furthermore, our data as well as recent work in Drosophila84 suggest that, in the context of developmental boundaries, one major role for architectural protein clusters (e.g. CTCF) is to robustly insulate the regulatory domains of major developmental genes and prevent spurious contacts with enhancers and/or silencers84. Accordingly, global 3D genome organization studies show that developmental genes and their cognate enhancers are typically located within the same TADs, suggesting a strong functional overlap between topological and regulatory domains14. Nevertheless, there are loci, with the Hox gene clusters being a notable example, in which genes and their cognate enhancers can be located in different TADs30,32,97. In these cases, the location of genes within or close to TAD boundaries might allow them to bypass those boundaries and communicate with enhancers located in more than one TAD in order to achieve proper gene expression patterns31,32,36,98. In contrast, for the two developmental genes that we investigated, we found no evidences indicating that CTCF clusters facilitate their expression or the communication with their cognate enhancers. Instead, we found that the sequential organization of genes and CTCF clusters at developmental boundaries is a prevalent and evolutionary conserved feature that seems to strengthen the insulation of important developmental loci.

Lastly, our findings might also have relevant medical implications, particularly in the context of structural variants (SV) causing congenital defects through long-range pathomechanisms42. For example, deletions spanning TAD boundaries can lead to pathological gains in gene expression through enhancer adoption mechanisms whereby enhancers can induce the expression of non-target genes42,99. Our data indicates that the pathological effects of this type of SVs might change (i.e. stronger or weaker induction of non-target genes) depending on whether the deletions include not only CTCF sites, but also nearby developmental genes.

Methods

All the experiments presented in this work comply with all relevant ethical and biosafety regulations and have been approved by the IBBTEC Biosafety commission and the CSIC Ethics committee.

ESC maintenance and differentiation protocol

E14Tg2a (E14) mouse ESC (a kind gift from Joanna Wysocka’s lab (Standford University) were cultured on gelatin-coated plates using Knock-out DMEM (Life Technologies, 10829018) supplemented with 15% FBS (Life Technologies, 10082147), leukemia inhibitory factor (LIF), antifungal and antibiotics (Sigma-Aldrich, A5955), β-mercaptoethanol (ThermoFisher Scientific, 21985023), Glutamax (ThermoFisher Scientific, 35050038) and MEM NEAA (ThermoFisher Scientific, 11140035). Cells were cultured at 37 °C with 5% CO2.

For the NPC differentiation63, ESCs were plated at 20,000 cells/cm2 on geltrex-coated plates (ThermoFisher, A1413302) and grown in N2B27 medium: Advanced Dulbecco’s Modified Eagle Medium F12 (Life Technologies, 21041025) and Neurobasal medium (Life Technologies, 12348017) (1:1), supplemented with 1 × N2 (R&D Systems, AR009), 1 × B27 (Life Technologies, 12587010), 2 mM l-glutamine (Life Technologies, 25030024) and 0.1 mM 2-mercaptoethanol (Life Technologies, 31350010)). During the six days of differentiation, the N2B27 medium was additionally supplemented with the following components: bFGF (ThermoFisher Scientific, PHG0368) 10 ng/ml from day 0 to day 2, Xav939 (Sigma-Aldrich, X3004-5MG) 5 µM from day 2 to day 6, BSA (ThermoFisher Scientific, 15260037) 1 mg/ml at day 0 and 40 µg/mL the remaining days.

RNA isolation, cDNA synthesis and RT-qPCR

Total RNA was isolated using the NZY Total RNA Isolation kit (NZYTech, MB13402) following the manufacturer’s instructions. RNA was reverse transcribed into cDNA using the NZY First-Strand cDNA Synthesis Kit (NZYTech, MB13402). For each reaction, 1 µg of RNA was incubated with 10 µL of NZYRT 2x Master Mix, 2 µL of NZYRT Enzyme Mix and nuclease-free water to a total volume of 20 µL at 25 °C for 10 min, followed by 30 min at 50 °C. The enzyme was heat inactivated at 85 °C for 5 min. To digest the remaining RNA, 1 µL of NZY RnaseH was added to the reaction and incubated at 37 °C for 20 min.

RT-qPCRs were performed using the CFX 384 detection system (Bio-Rad) using NZYSpeedy qPCR Green Master Mix (2x) (NZYtech, MB224). For each sample, RT-qPCRs were performed as technical triplicates using the primers listed in Data S3. For each cell line, the number of clonal lines and biological replicates analysed in each case is indicated in the corresponding figure legends. For the investigated genes (i.e. Gbx2, Asb18, Six3 and Six2), gene expression fold-changes between each cell line and the corresponding WT cells were calculated using the 2−ΔΔCT method, using Eef1a1 and Hprt1 as housekeeping genes. Fold-changes are shown as Log2 values and are plotted using box plots in which (i) the upper and lower parts of the box are the upper and lower quartiles, respectively, (ii) the horizontal line that splits the box in two is the median and (iii) the upper and lower whiskers indicate the maximum and minimum, respectively.

CRISPR-Cas9 genome editing

The design of the CRISPR/Cas9 guide sequences (gRNA) was performed using the CRISPR Benchling software tool (https://www.benchling.com/crispr/) (Data S3). In order to increase the cutting efficiency of the gRNA, a guanine nucleotide was added at the first position of the sequence and a restriction site for the BbsI enzyme (R0539L, NEB) was added to the beginning of the gRNA for cloning purposes. For each sgRNA, two oligonucleotides were synthesized (Sigma), annealed and cloned into a CRISPR-Cas9 expression vector (pX330-hCas9-long-chimeric-grna-g2p; gift from Leo Kurian’s laboratory). The hybridized oligos were cloned into the pX330-hCas9-long-chimeric-grna-g2p using 50 ng of BbsI-digested vectors and 1 µl ligase (Thermo Fisher, EL0013) in a total volume of 20 μl. The ligation reaction was incubated for 1 h at room temperature. For each cell line, ESC were transfected with the corresponding pair of gRNAs-Cas9 expressing vectors using Lipofectamine following manufacturer’s recommendations (Thermo Scientific, L3000001). After 24 h, transfected cells were selected by treating them with puromycin for 48 h. Single-cell isolation of surviving cells was performed by serial dilution and seeding in 96-well plates. Next, clones with the desired genetic rearrangements (i.e. deletions or inversions) were identified by PCR using the primers listed in Data S3. DNA extraction was performed using Lysis Buffer: 25 mM KCl (SigmaAldrich, 27810.295), 5 mM TRIS (Sigma-Aldrich, 0497-5KG) pH8.3, 1.25 mM MgCl2 (VWR BDH7899-1), 0.225% IGEPAL (Sigma-Aldrich, I8896-50 ML) and 0.225% Tween20 (VWR). Proteinase K (ThermoFisher Scientific, EO0492) was added to a final concentration of 0.4 µg/µl before use.

Using E14Tg2a mESC, the following cell lines were generated in this work:

Gbx2/Asb18 locus: Δ3xCTCF, 71 Kb INV, Δ3xCTCF:71 Kb INV, ΔProm Gbx2, Δ3xCTCF:ΔProm Gbx2, ΔCTCF SE Gbx2, Gbx2 INV, Gbx2 INV:Δ3xCTCF

Six3/Six2 locus: Δ6xCTCF, 156 Kb INV, Δ6xCTCF:156 Kb INV, Six3−/−, Δ6xCTCF:Six3−/−, Δ4xCTCF, Δ4xCTCF:Six3−/−, Δ6xCTCF:Six2−/−, 226 Kb INV

ChIP-Seq

~4 × 107 cells were used for RAD21 (abcam, ab154769) and ~1 × 107 cells for H3K27ac (Active Motif, 39133). Cells were crosslinked with 1% formaldehyde for 10 min at room temperature (RT) and quenched with 0.125 M glycine for 10 min. Cells were consecutively incubated with three different lysis buffers (Buffer 1: 50 mM HEPES, 140 mM NaCl, 1 mM EDTA, 10% glycerol, 0.5% NP-40, 0.25% TX-100; Buffer 2: 10 mM Tris, 200 mM NaCl, 1 mM EDTA, 0.5 mM EGTA; Buffer 3: 10 mM Tris, 100 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 0.1% Na-deoxycholate, 0.5% N-lauroylsarcosine) in order to isolate chromatin. Next, chromatin was sonicated for 8 min in Buffer 3 (20 s on, 30 s off, 25% amplitude) using an EpiShear probe sonicator (Active Motif).

The sonicated chromatin was mixed with 3 μg of H3K27ac antibody or 10 μg of Rad21 antibody and incubated overnight at 4 °C. Next, 50−100 μl of Dynabeads (Thermo Fisher, 10-002-D) were aliquoted into a microtube for each ChIP reaction. The dynabeads were washed with 1 ml of cold blocking solution (0,5% BSA and 1x PBS). The antibody bound chromatin was added to the washed beads and incubated for 4 h at 4 °C. Magnetic beads were washed five times with RIPA buffer (50 mM Hepes, 500 mM LiCl, 1 mM EDTA, 1% NP-40 and 0,7% Na-Deoxycholate). The chromatin was eluted and incubated at 65 °C overnight to reverse the crosslinking. Next, samples were treated with 0.2 mg/ml Rnase A and 0.2 mg/ml proteinase K. DNA purification was performed using Zymoclean Gel DNA Recovery kit (Zymo Research, D4008).

Regarding the computational processing of the ChIP-seq data, reads were subject to quality control and trimming of low quality regions and/or adapters using fastqc (https://www.bioinformatics.babraham.ac.uk/projects/fastqc/), MultiQC100 and trimmomatic101. Next, reads were mapped to mm10 reference genome with Bowtie2102. After read mapping, only reads with a mapping quality above 10 were kept and duplicated reads were removed with SAMtools103. Afterwards, bigwig files were generated with bamCoverage from deepTools104 applying the reads per genome coverage normalization.

Capture-C

Capture-C experiments were performed as previously described105. 5 × 106 cells were crosslinked with 2% formaldehyde for 10 min and quenched with 0.125 M glycine for 10 min. Cells were washed with PBS and resuspended in lysis buffer (10 mM Tris pH 8, 10 mM NaCl, 0.2% NP-40 and 1× protease inhibitors) during 20 min on ice. Following centrifugation, the pellet was resuspended in 215 µL 1×CutSmart buffer and transferred to a microcentrifuge tube. The resuspended pellet was mixed with 60 µl 10× CutSmart buffer, 393.5 µl water and 9.5 µl 20% (vol/vol) SDS (0.28% final concentration) (Invitrogen, cat. no. AM9820) followed by 1 h incubation at 37 °C while shaking on a thermomixer at 500 rpm (intermittent shaking: 30 s on/30 s off). Then, 20% vol/vol Triton X 100 was added at a final concentration of 1.67% vol/vol followed by another incubation at 37 °C for 1 h while shaking. Chromatin was digested by adding 25 µL NlaIII (250 U, R0125L) and incubating at 37 °C for several hours, followed by addition of another 25 µL of NlaIII and incubation at 37 °C overnight. The digested chromatin was ligated with 8 µl (240U) of T4 DNA ligase (Life Tech, cat. no. EL0013) for 18 h at 16 °C. Samples were treated with Proteinase K (3U; Thermo Fisher, cat. no. EO0491) and RNase A (7.5 mU; Roche: 1119915), and DNA was purified using the Qiagen kit (28506). After checking the quality of the digestion and subsequent ligation, chromatin was sonicated for 30 cycles (30 s on, 30 s off, 25% amplitude) using an EpiShear probe sonicator (Active Motif) and DNA samples were purified using AMPure XP SPRI beads (Beckman Coulter, cat. no. A63881). Libraries were prepared using the NEBNext Ultra II kit (New England Biolabs, cat. no. E7645S/L). Index primers set 1 and 2 from the NEBNext Multiplex Oligos for Illumina kit (New England Biolabs, E7335S/L E7500S/L) were incorporated using Herculase II Fusion Polymerase Kit (Agilent, cat. no. 600677). The resulting libraries were pooled (six libraries/pool) and a double Hybridization capture using ssDNA probes was performed following a modified version of the Roche HyperCapture streptavidin pull-down protocol (Roche, cat. no. 09075763001) described in ref. 105. Libraries were sequenced using Novaseq6000_150PE_2,25 Gb/lib (15 Mreads/lib). For each of the investigated cell lines, Capture-C experiments were performed as two independent biological replicates.

Capsequm2 (http://apps.molbiol.ox.ac.uk/CaptureC/cgi-bin/CapSequm.cgi) was used to design the ssDNA probes (120 bp probe length). The sequences of the ssDNA probes are listed in Data S3.

Analysis and quantification of Capture-C data

Capture C reads were subject to quality control and trimming of low quality regions and/or adapters using fastqc (https://www.bioinformatics.babraham.ac.uk/projects/fastqc/), MultiQC100 and trimmomatic101. Next, the reads were processed with capC-MAP106, considering the restriction enzyme NlaIII (cutting site CATG), the mm10 reference genome and normalize = TRUE. The coordinates (mm10) of the viewpoints (i.e. «targets» according to capC-MAP terminology) were:

Gbx2 SE: chr1 89869929 89870764

Six3 SE: chr17 85484699 85485038

Asb18 TSS: chr1 90013014 90013383

Six2 TSS: chr17 85688402 85689017

After running capC-MAP, the normalized pileup bedgraph files for intra-chromosomal contacts for each viewpoint were collected. According to capC-MAP documentation, the number of piled-up interactions per restriction fragment are normalized to reads per million, so that the sum of the number of reads associated to each viewpoint genome-wide is equal to one million. Subsequently, for the restriction sites without any detected interactions, a signal equal to 0 was assigned. In addition, only data from the regions chr1 89228752 90664659 (Gbx2-Asb18 locus) and chr17 84890284 86289254 (Six3-Six2 locus) was considered. Next, the bedgraph files of both replicates were averaged and the resulting bedgraph was converted to a bigwig with the usage of the bedGraphToBigWig UCSC tool107. Bigwig subtraction tracks were generated with the usage of bigwigCompare from deepTools104. For visualization purposes in the UCSC browser, the subtraction tracks were subject to smoothing (16 pixels window) in order to minimize the contribution of single restriction site fragments.

Annotation of developmental and housekeeping genes

Developmental genes were annotated based on the presence of broad H3K27me3/PcG domains around their TSS60,62 using a previously described strategy61,63. Briefly, H3K27me3 Chip-Seq fastq files from hESC (GSE24447; H3K27me3: SRR067372, input: SRR067371) and mESC (GSE89209; H3K27me3:SRR4453259, input: SRR4453262) were downloaded. Then, reads were mapped against mm10 and hg19 genomes with bowtie2 and peaks were called with MACS considering the broad peak calling mode108. After peak calling, only peaks with a fold enrichment >3 and q value < 0.1 were kept. Next, peaks within 1 Kb of each other were merged using bedtools, and associated with a protein coding gene when overlapping a TSS. Subsequently, the knee of the peak size distribution was evaluated with findiplist() (inflection R package; https://cran.r-project.org/web/packages/inflection/vignettes/inflection.html). Upon curvature analyses, genes associated with a H3K27me3 peak>7Kb were defined as developmental genes (Mouse: n = 967; Human: n = 1045).

For the annotation of housekeeping genes, one list for humans and one for mice (Mouse: n = 3277; Human: n = 2176) were obtained from the Housekeeping and Reference Transcript Atlas database109.

For the analyses presented in Fig. S5, developmental genes were defined as those associated with the Gene Ontology (GO) term developmental process; (GO:0032502) using AmiGO110. These developmental genes were further sub-divided in two groups based on the presence or absence of broad H3K27me3/PcG domains around their TSS as described above.

In silico analysis of TAD gene density

TAD maps previously generated in 11 mouse and 37 human cell types were considered. To ensure consistency in TAD calling, the genomic coordinates of all TADs and their boundaries were retrieved from the 3D Genome Browser database111, in which TAD boundaries were consistently called using the directionality index approach initially established by Dixon et al.13. Regarding human TAD maps, the 37 hg19 TAD maps available in the 3D Genome browser were used111. With respect to mice, 11 TAD maps were used: (i) eight mm10 TAD maps available in the 3D Genome browser111, (ii) three mm9 TAD maps (CH12_Lieberman-raw_TADs.txt, cortex_Dixon2012-raw_TADs.txt and mESC_Dixon2012-raw_TADs.txt), also available in 3D genome browser and that were liftover to mm10 using the UCSC liftover tool112. For the eight mm10 TAD maps from the 3D Genome browser, with the exception of the files G1E-ER4.rep1-raw.domains and G1E-ER4.rep2-raw.domains, the prefix chr was added to the chromosome name, chr23 was relabeled to chrX and chr24 to chrY.

For each TAD, gene density was computed as the number of TSSs (based on hg19 and mm10 RefSeq curated annotations downloaded through the UCSC Table browser113) located within each TAD divided by the length of the TAD. Subsequently, for each TAD map, the TADs were sorted based on increasing gene densities, and three groups of TADs of equal size were considered: low density (LD), medium density (MD) and high density (HD) TADs. Next, for each TAD map, we computed whether developmental genes were over represented among the genes found within LD TADs using a Fisher test. Lastly, Gene Ontology functional enrichment analyses were performed for two different TAD maps (hESC Dixon and mESC Dixon, available in 3D genome browser) using the WebGestalt R package114 and considering the genes located in the three different groups of TADs (LD, MD and HD) described above. The WebGestalt functional enrichment analysis were performed using the ORA (over representation analyses) method and all genes as the reference gene list (group of genes used to compare and compute enrichments).

In silico analysis of gene distribution within TADs

When computing gene distribution within TADs, the TSSs of genes were taken as a reference. TSS coordinates were obtained from RefSeq curated annotation (downloaded through the UCSC Table browser113) for both human and mice. Gene distribution within TADs was computed in multiple TAD maps independently (37 TAD maps in humans and 11 in mice, see previous methods section).

Each TAD was divided in ten bins of equal sizes. Therefore, the size of each bin is 10% of the size of its corresponding TAD. Regarding the labeling of the bins, when moving from the boundaries of the TAD (TAD start or end coordinates) towards its interior, the first bin was labeled as bin 1, the next one as bin 2 and so on, until reaching bin 5, which is the closest to the center of the TAD (Fig. 1B). The genomic regions located outside TADs (inter-TAD) were assigned to bin 0 (Fig. 1B). For each of the considered TAD maps, the TSSs of genes were assigned to bins attending to their location in the genome. Furthermore, for the analyses presented in Fig. S4, the distance between each TSS located in bin 1 and its closest TAD boundary was computed.

Global analysis of insulation strength and CTCF binding at TAD boundaries

The CTCF data used in these analyses were obtained from mESC (GSE36027, CTCF replicates: SRR489719 and SRR489720, input replicates: SRR489731 and SRR489732) and hESCs (GSE116862, CTCF replicates: SRR7506641 and SRR7506642, input replicates: SRR7506652 and SRR7506653). After downloading the corresponding fastq files, reads from both replicates were merged and subject to quality control and trimming of low quality regions and/or adapters using fastqc, MultiQC100 and trimmomatic101. Next, reads were mapped to either mm10 or hg19 genomes with Bowtie2102. After read mapping, only reads with a mapping quality above 10 were kept and duplicated reads were removed with SAMtools103. Afterwards, CTCF bigwig files were generated with bamCoverage from deepTools applying the reads per genome coverage normalization104. CTCF peaks were called with MACS2108 and only those peaks with a fold change > 4 and q value < 0.01 were considered. Lastly, using the coordinates of the CTCF peaks and the CTCF bigwig files, the maximum intensity of each CTCF peak was calculated using the bigWigAverageOverBed UCSC binary tool.

The insulation score and boundary strength datasets used for our analyses were obtained from different sources and through various procedures. Regarding mESC data, a bigwig file (file ID: 4DNFIMVJ2YV3) with mm10 insulation scores obtained from previously generated Hi-C data19 was downloaded from the 4D Nucleome data portal115. In addition, a bed file (file id: 4DNFI1S7FI1U) with boundary strength values as computed by cooltools116 for the bins mapped as TAD boundaries was also downloaded from the 4D Nucleome data portal115. Regarding hESC data, .hic files from two replicates (GSM3262956 and GSM326295720) were downloaded from GEO. Next, .hic files were converted to .cool format with the hic2cool software (https://github.com/4dn-dcic/hic2cool) considering the 10 Kb contact resolution matrix. Afterwards both replicates were merged with cooler merge and normalised with cooler balance117. Subsequently, insulation scores were computed with cooltools (based on the diamond insulation score technique) using a window size of 100 Kb. In addition a bigwig file was also created in this step with cooltools by specifying the –bigwig option. Moreover, boundary strength metrics were also computed by cooltools for the bins considered as TAD boundaries. In order to make these boundary strength values more comparable to those obtained from the 4D Nucleome data portal, only TAD boundaries with a strength larger than 0.2 were considered, while a strength value = 0 was assigned to the remaining boundaries

Once ready, the insulation score, boundary strength and CTCF datasets were used to compute several metrics for TAD boundaries in both mice and humans. TAD boundaries were defined using the start and end coordinates of TADs previously identified in mESC19 and hESC118. Next, each TAD boundary was expanded by ±50 Kb and the following metrics were calculated within the resulting 100 Kb window: (i) the number of overlapping CTCF peaks, (ii) the CTCF aggregated signal (sum of the CTCF bigwig signal for all the peaks overlapping with the 100 kb window), (iii) the insulation score (minimum insulation score value in the 100 kb window computed with bigWigAverageOverBed) and (iv) the boundary strength (maximum boundary strength value computed by cooltools for any bin in the 100 kb window). In addition, TAD boundaries were classified as Developmental or Other: Developmental boundaries were defined as those associated with a developmental gene located in bin 1 (Fig. 1 1B); Other included all the remaining boundaries. Moreover, a set of 5000 random TAD boundaries was generated by randomly selecting 5000 regions in the mouse and human genomes.

The chicken and zebrafish CTCF ChIP-seq profiles shown in Fig. S9 were previously generated in HH18 chicken embryos (GSM5835469)119 and 48 hpf zebrafish embryos (GSM5344494)120, respectively.

In silico analysis of the distribution of CTCF sites and CTCF motif orientation around genes located close to TAD boundaries

Gene located close to TAD boundaries were defined as those assigned to bin 1 (Fig. 1B) according to TAD maps previously generated in mESC19 and hESC118. Then, for each bin 1 gene, the number of CTCF peaks (obtained as described in the previous section) located within a ±100 Kb window around its TSS was calculated. The 100 Kb window extending from the gene TSS towards the TAD boundary was defined as the outer window, while the 100 Kb window extending from the gene TSS towards the center of the TAD was defined as the inner window (Fig. 2C). After calculating the number of CTCF peaks in the inner and outer windows, we computed the ΔCTCFpeaks@Bdry metric for each gene as the number of CTCF peaks located in the inner window minus the number of CTCF peaks located in the outer window. In addition, the CTCF bigwig signals associated with the CTCF peaks were used to calculate the ΔCTCFsignal@Bdry metric for each gene as the aggregated signal for all the CTCF peaks located in the inner window minus the aggregated signal for all the CTCF peaks located in the outer window.

In addition, for the CTCF peaks located in the inner and outer windows of bin 1 genes described above, their motif orientation was obtained using the CTCF package (https://github.com/dozmorovlab/CTCF). Briefly, JASPAR 2022 CTCF motif predictions for CTCF motif MA0139.1 were considered: (i) mm10.JASPAR2022_CORE_vertebrates_non_redundant_v2 data for mouse analyses, and (ii) hg19.JASPAR2022_CORE_vertebrates_non_redundant_v2 data for human analyses. First, the CTCF peaks were intersected with the motif coordinates (as provided by the CTCF package) using bedtools. Next, for each CTCF peak, the orientation associated with the overlapping CTCF motif showing the lowest q-value was defined as the CTCF peak orientation. Finally, taking into account the position of the center of the TADs, the CTCF peaks orientation was defined as +1 when oriented towards the center of the TAD (inward site) or as −1 when oriented away from the center of the TAD (outward site). To compare the distribution of inward and outward CTCF sites between the inner and outer window of the different bin1 gene categories (i.e. All, Developmental and Housekeeping), Chi-squared tests were calculated. The Cramér’s V effect size estimator121 was calculated with rcompanion.

Analysis of RNA Pol2 Pausing Index (PI)

The pausing index (PI) was calculated as the ratio of the average PRO-Seq signal (from mESC69) or GRO-Seq (from hiPSC70) between the gene promoter (from 50 bp upstream of the TSS to 250 bp downstream of the TSS) and the gene body (from 250 bp downstream of the TSS to the Transcription Termination Site (TTS)). TSS and TTS coordinates were obtained from RefSeq curated annotation, as described above.

For mouse analyses, strand-specific PRO-Seq signal bedgraph files from mESC were retrieved from GEO (GSE178230). The downloaded files were GSE178230_mm10_mESC_PROseq_mESC_N4_F.bedGraph and GSE178230_mm10_mESC_PROseq_mESC_N4_R.bedGraph. Bedgraph files were converted to bigwigs with the bedGraphToBigWig tool. Next, for each transcript, the average bigwig signal for the promoter and gene body regions were obtained with the bigWigAverageOverBed tool and used for the PI calculation. For each transcript, only the PRO-Seq data of its corresponding strand was considered.

For human analyses, GRO-Seq bedgraph files from induced pluripotent stem cells (iPSCs), derived of two different donors, were retrieved from GEO (GSE117086). The downloaded files were: GSM3271001_iPSC_11104_A.bedGraph and GSM3271002_iPSC_11104_B.bedGraph. Next, the two bedgraph files were combined and converted to a single bigwig with the usage of the bedGraphToBigWig and bigWigMerge UCSC binary tools. Subsequently, for each transcript, the average bigwig signals for the promoter and gene body regions were obtained with bigWigAverageOverBed tool and used for the PI calculation.

After PIs calculation, transcripts with not finite PIs were discarded with the usage of is.finite() R function. When filtering genes by gene expression levels, those with expression levels above 5 FPKM for mESC58 or 5 RPKM for hESC (GSE24447, file GSM602289_ESC_RPKM.txt) were considered to be active. When selecting genes based on their proximity to TAD boundaries, only transcripts whose TSS were located within bin 1 (Fig. 1B) according to mESC or hESC TAD maps (see previous Method sections) were considered. In all the comparisons, for genes with multiple transcripts, the highest pausing index computed for all the transcripts was kept as a reference.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

Supplementary information

Peer Review File(1.4M, pdf)

Source data

Source Data(124K, xlsx)

Acknowledgements

We would like to thank all the Rada-Iglesias lab members for insightful comments and suggestions. Work in the Rada-Iglesias laboratory is supported by grant PID2021-123030NB-I00 (AR-I) funded by MCIN/AEI/ 10.13039/501100011033 and by ERDF A way of making Europe, grant RED2022-134100-T (AR-I) (REDEVNEURAL 3.0) funded by MCIN/AEI /10.13039/501100011033, grant ERC CoG PoisedLogic (862022) (A.R.-I.) funded by the European Research Council, grant ENHPATHY H2020-MSCA-ITN-2019-860002 (A.R.-I.) funded by the European Commission, grant Chrom_rare HORIZON-MSCA-2021-DN-01-101073334 (A.R.-I.) funded by the European Commission. E.M.B was granted with a fellowship from the network PIE-202120E047-Conexiones-Life.

Author contributions

T.E. performed most of the experimental studies. V.S-G. carried out the computational analysis and contributed to the conceptual design of the project. P.R., M.M-S. and E.M-B. participated in the generation some of the transgenic mESC lines. E.H. contributed to the design of the project and supervised the experimental work. A.R-I. supervised the work. T.E., V.S-G. and A.R.-I. wrote the manuscript and prepared the figures.

Peer review

Peer review information

Nature Communications thanks the anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available.

Data availability

All the generated ChIP-seq and Capture-C data are publically available through GEO under accession codes: GSE252218, GSE252080. The following publically available and previously generated datasets were used in this study: GSE24447, GSE89209, GSE36027, GSE116862, GSE178230, GSE117086Source data are provided with this paper.

Competing interests

The authors declare no competing interests.

Inclusion and ethics

The authors have adhered to the inclusion & ethics standards of the Nature portfolio journals.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

These authors contributed equally: Thais Ealo, Victor Sanchez-Gaya.

Contributor Information

Victor Sanchez-Gaya, se.nacinu@ayagzehcnas.rotciv.

Endika Haro, se.nacinu@orah.akidne.

Alvaro Rada-Iglesias, se.nacinu@adar.oravla.

Supplementary information

The online version contains supplementary material available at 10.1038/s41467-024-51602-4.

References

1. Furlong, E. E. M. & Levine, M. Developmental enhancers and chromosome topology. Science361, 1341–1345 (2018). 10.1126/science.aau0320 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
2. Lagha, M., Bothma, J. P. & Levine, M. Mechanisms of transcriptional precision in animal development. Trends Genet.28, 409–416 (2012). 10.1016/j.tig.2012.03.006 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
3. Long, H. K., Prescott, S. L. & Wysocka, J. Ever-changing landscapes: transcriptional enhancers in development and evolution. Cell167, 1170–1187 (2016). 10.1016/j.cell.2016.09.018 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. West, A. G., Gaszner, M. & Felsenfeld, G. Insulators: many functions, many mechanisms. Genes Dev.16, 271–288 (2002). 10.1101/gad.954702 [Abstract] [CrossRef] [Google Scholar]
5. Hnisz, D., Day, D. S. & Young, R. A. Insulated neighborhoods: structural and functional units of mammalian gene control. Cell167, 1188–1200 (2016). 10.1016/j.cell.2016.10.024 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
6. Bell, A. C., West, A. G. & Felsenfeld, G. The protein CTCF is required for the enhancer blocking activity of vertebrate insulators. Cell98, 387–396 (1999). 10.1016/S0092-8674(00)81967-4 [Abstract] [CrossRef] [Google Scholar]
7. Geyer, P. K. & Corces, V. G. DNA position-specific repression of transcription by a Drosophila zinc finger protein. Genes Dev6, 1865–1873 (1992). 10.1101/gad.6.10.1865 [Abstract] [CrossRef] [Google Scholar]
8. Cavalheiro, G. R. et al. CTCF, BEAF-32, and CP190 are not required for the establishment of TADs in early Drosophila embryos but have locus-specific roles. Sci. Adv.9, eade1085 (2023). 10.1126/sciadv.ade1085 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
9. Chathoth, K. T. et al. The role of insulators and transcription in 3D chromatin organization of flies. Genome Res.32, 682–698 (2022). 10.1101/gr.275809.121 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
10. Kaushal, A. et al. Essential role of Cp190 in physical and regulatory boundary formation. Sci. Adv.8, eabl8834 (2022). 10.1126/sciadv.abl8834 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
11. Nora, E. P. et al. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell169, 930–944.e22 (2017). 10.1016/j.cell.2017.05.004 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
12. Ohlsson, R., Renkawitz, R. & Lobanenkov, V. CTCF is a uniquely versatile transcription regulator linked to epigenetics and disease. Trends Genet17, 520–527 (2001). 10.1016/S0168-9525(01)02366-6 [Abstract] [CrossRef] [Google Scholar]
13. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature485, 376–380 (2012). 10.1038/nature11082 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
14. Harmston, N. et al. Topologically associating domains are ancient features that coincide with Metazoan clusters of extreme noncoding conservation. Nat. Commun.8, 441 (2017). 10.1038/s41467-017-00524-5 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
15. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell171, 305–320.e24 (2017). 10.1016/j.cell.2017.09.026 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
16. Schwarzer, W. et al. Two independent modes of chromatin organization revealed by cohesin removal. Nature551, 51–56 (2017). 10.1038/nature24281 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
17. Rowley, M. J. et al. Evolutionarily conserved principles predict 3D chromatin organization. Mol. Cell67, 837–852.e7 (2017). 10.1016/j.molcel.2017.07.022 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
18. Kaushal, A. et al. CTCF loss has limited effects on global genome architecture in Drosophila despite critical regulatory functions. Nat. Commun.12, 1011 (2021). 10.1038/s41467-021-21366-2 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
19. Bonev, B. et al. Multiscale 3D genome rewiring during mouse neural development. Cell171, 557–572.e24 (2017). 10.1016/j.cell.2017.09.043 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
20. Zhang, Y. et al. Transcriptionally active HERV-H retrotransposons demarcate topologically associating domains in human pluripotent stem cells. Nat. Genet.51, 1380–1388 (2019). 10.1038/s41588-019-0479-7 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
21. Zhang, S. et al. RNA polymerase II is required for spatial chromatin reorganization following exit from mitosis. Sci. Adv.7, eabg8205 (2021). 10.1126/sciadv.abg8205 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
22. Zhang, S., Übelmesser, N., Barbieri, M. & Papantonis, A. Enhancer-promoter contact formation requires RNAPII and antagonizes loop extrusion. Nat. Genet.55, 832–840 (2023). 10.1038/s41588-023-01364-4 [Abstract] [CrossRef] [Google Scholar]
23. Banigan, E. J. et al. Transcription shapes 3D chromatin organization by interacting with loop extrusion. Proc. Natl Acad. Sci. USA.120, e2210480120 (2023). 10.1073/pnas.2210480120 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
24. Bozhilov, Y. K. et al. A gain-of-function single nucleotide variant creates a new promoter which acts as an orientation-dependent enhancer-blocker. Nat. Commun.12, 3806 (2021). 10.1038/s41467-021-23980-6 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
25. Barshad, G. et al. RNA polymerase II dynamics shape enhancer-promoter interactions. Nat. Genet.55, 1370–1380 (2023). 10.1038/s41588-023-01442-7 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
26. Zhang, D. et al. Alteration of genome folding via contact domain boundary insertion. Nat. Genet.52, 1076–1087 (2020). 10.1038/s41588-020-0680-8 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
27. Hsieh, T.-H. S. et al. Resolving the 3D landscape of transcription-linked mammalian chromatin folding. Mol. Cell78, 539–553.e8 (2020). 10.1016/j.molcel.2020.03.002 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
28. Zhang, H. et al. CTCF and transcription influence chromatin structure re-configuration after mitosis. Nat. Commun.12, 5157 (2021). 10.1038/s41467-021-25418-5 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Jiang, Y. et al. Genome-wide analyses of chromatin interactions after the loss of Pol I, Pol II, and Pol III. Genome Biol.21, 158 (2020). 10.1186/s13059-020-02067-3 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
30. Beccari, L. et al. Dbx2 regulation in limbs suggests interTAD sharing of enhancers. Dev. Dyn.250, 1280–1299 (2021). 10.1002/dvdy.303 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
31. Luppino, J. M. et al. Cohesin promotes stochastic domain intermingling to ensure proper regulation of boundary-proximal genes. Nat. Genet.52, 840–848 (2020). 10.1038/s41588-020-0647-9 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
32. Hung, T.-C., Kingsley, D. M. & Boettiger, A. N. Boundary stacking interactions enable cross-TAD enhancer-promoter communication during limb development. Nat. Genet.56, 306–314 (2024). 10.1038/s41588-023-01641-2 [Abstract] [CrossRef] [Google Scholar]
33. Chakraborty, S. et al. Enhancer–promoter interactions can bypass CTCF-mediated boundaries and contribute to phenotypic robustness. Nat. Genet.55, 280–290 (2023). 10.1038/s41588-022-01295-6 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
34. Balasubramanian, D. et al. Enhancer–promoter interactions can form independently of genomic distance and be functional across TAD boundaries. Nucleic Acids Res. gkad1183. 10.1093/nar/gkad1183 (2023). [Europe PMC free article] [Abstract]
35. Chang, L.-H., Ghosh, S. & Noordermeer, D. TADs and their borders: free movement or building a wall? J. Mol. Biol.432, 643–652 (2020). 10.1016/j.jmb.2019.11.025 [Abstract] [CrossRef] [Google Scholar]
36. Chang, L.-H. et al. Multi-feature clustering of CTCF binding creates robustness for loop extrusion blocking and Topologically Associating Domain boundaries. Nat. Commun.14, 5615 (2023). 10.1038/s41467-023-41265-y [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
37. Vian, L. et al. The energetics and physiological impact of cohesin extrusion. Cell173, 1165–1178.e20 (2018). 10.1016/j.cell.2018.03.072 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
38. Kubo, N. et al. Promoter-proximal CTCF binding promotes distal enhancer-dependent gene activation. Nat. Struct. Mol. Biol.28, 152–161 (2021). 10.1038/s41594-020-00539-5 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
39. Paliou, C. et al. Preformed chromatin topology assists transcriptional robustness of Shh during limb development. Proc. Natl Acad. Sci.116, 12390–12399 (2019). 10.1073/pnas.1900672116 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
40. Long, H. K. et al. Loss of extreme long-range enhancers in human neural crest drives a craniofacial disorder. Cell Stem Cell27, 765–783.e14 (2020). 10.1016/j.stem.2020.09.001 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
41. Rinzema, N. J. et al. Building regulatory landscapes reveals that an enhancer can recruit cohesin to create contact domains, engage CTCF sites and activate distant genes. Nat. Struct. Mol. Biol.29, 563–574 (2022). 10.1038/s41594-022-00787-7 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
42. Spielmann, M., Lupiáñez, D. G. & Mundlos, S. Structural variation in the 3D genome. Nat. Rev. Genet.19, 453–467 (2018). 10.1038/s41576-018-0007-0 [Abstract] [CrossRef] [Google Scholar]
43. Harrold, C. L. et al. A functional overlap between actively transcribed genes and chromatin boundary elements. 10.1101/2020.07.01.182089 (2020).
44. Bartman, C. R., Hsu, S. C., Hsiung, C. C.-S., Raj, A. & Blobel, G. A. Enhancer regulation of transcriptional bursting parameters revealed by forced chromatin looping. Mol. Cell62, 237–247 (2016). 10.1016/j.molcel.2016.03.007 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
45. Lower, K. M. et al. Adventitious changes in long-range gene expression caused by polymorphic structural variation and promoter competition. Proc. Natl Acad. Sci. USA106, 21771–21776 (2009). 10.1073/pnas.0909331106 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
46. Topfer, S. K. et al. Disrupting the adult globin promoter alleviates promoter competition and reactivates fetal globin gene expression. Blood139, 2107–2118 (2022). 10.1182/blood.2021014205 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
47. Ohtsuki, S., Levine, M. & Cai, H. N. Different core promoters possess distinct regulatory activities in the Drosophila embryo. Genes Dev.12, 547–556 (1998). 10.1101/gad.12.4.547 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
48. Ohtsuki, S. & Levine, M. GAGA mediates the enhancer blocking activity of the eve promoter in the Drosophila embryo. Genes Dev.12, 3325–3330 (1998). 10.1101/gad.12.21.3325 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
49. Cai, H. N., Zhang, Z., Adams, J. R. & Shen, P. Genomic context modulates insulator activity through promoter competition. Development128, 4339–4347 (2001). 10.1242/dev.128.21.4339 [Abstract] [CrossRef] [Google Scholar]
50. Palstra, R., De Laat, W. & Grosveld, F. Chapter 4 β‐globin regulation and long‐range interactions. in Advances in Genetics. Vol. 61, 107–142 (Elsevier, 2008). [Abstract]
51. Oudelaar, A. M. et al. A revised model for promoter competition based on multi-way chromatin interactions at the α-globin locus. Nat. Commun.10, 5412 (2019). 10.1038/s41467-019-13404-x [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
52. Fukaya, T., Lim, B. & Levine, M. Enhancer control of transcriptional bursting. Cell166, 358–368 (2016). 10.1016/j.cell.2016.05.025 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
53. Allahyar, A. et al. Enhancer hubs and loop collisions identified from single-allele topologies. Nat. Genet.50, 1151–1160 (2018). 10.1038/s41588-018-0161-5 [Abstract] [CrossRef] [Google Scholar]
54. Barlow, D. P. Competition—a common motif for the imprinting mechanism? EMBO J.16, 6899–6905 (1997). 10.1093/emboj/16.23.6899 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
55. Lim, B. & Levine, M. S. Enhancer-promoter communication: hubs or loops? Curr. Opin. Genet. Dev.67, 5–9 (2021). 10.1016/j.gde.2020.10.001 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
56. Deng, H. & Lim, B. Shared transcriptional machinery at homologous alleles leads to reduced transcription in early Drosophila embryos. Front. Cell Dev. Biol.10, 912838 (2022). 10.3389/fcell.2022.912838 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
57. Sabi, R. & Tuller, T. Modelling and measuring intracellular competition for finite resources during gene expression. J. R. Soc. Interface16, 20180887 (2019). 10.1098/rsif.2018.0887 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
58. Cruz-Molina, S. et al. PRC2 facilitates the regulatory topology required for poised enhancer function during pluripotent stem cell differentiation. Cell Stem Cell20, 689–705.e9 (2017). 10.1016/j.stem.2017.02.004 [Abstract] [CrossRef] [Google Scholar]
59. Laugsch, M. et al. Modeling the pathological long-range regulatory effects of human structural variation with patient-specific hiPSCs. Cell Stem Cell24, 736–752.e12 (2019). 10.1016/j.stem.2019.03.004 [Abstract] [CrossRef] [Google Scholar]
60. Rehimi, R. et al. Epigenomics-based identification of major cell identity regulators within heterogeneous cell populations. Cell Rep.17, 3062–3076 (2016). 10.1016/j.celrep.2016.11.046 [Abstract] [CrossRef] [Google Scholar]
61. Sánchez-Gaya, V. & Rada-Iglesias, A. POSTRE: a tool to predict the pathological effects of human structural variants. Nucleic Acids Res51, e54 (2023). 10.1093/nar/gkad225 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
62. Shim, W. J. et al. Conserved epigenetic regulatory logic infers genes governing cell identity. Cell Syst.11, 625–639.e13 (2020). 10.1016/j.cels.2020.11.001 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
63. Pachano, T. et al. Orphan CpG islands amplify poised enhancer regulatory activity and determine target gene responsiveness. Nat. Genet.53, 1036–1049 (2021). 10.1038/s41588-021-00888-x [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
64. Kraft, K. et al. Serial genomic inversions induce tissue-specific architectural stripes, gene misexpression and congenital malformations. Nat. Cell Biol.21, 305–310 (2019). 10.1038/s41556-019-0273-x [Abstract] [CrossRef] [Google Scholar]
65. Lenhard, B., Sandelin, A. & Carninci, P. Metazoan promoters: emerging characteristics and insights into transcriptional regulation. Nat. Rev. Genet.13, 233–245 (2012). 10.1038/nrg3163 [Abstract] [CrossRef] [Google Scholar]
66. Yan, K.-K., Lou, S. & Gerstein, M. MrTADFinder: a network modularity based approach to identify topologically associating domains in multiple resolutions. PLoS Comput. Biol.13, e1005647 (2017). 10.1371/journal.pcbi.1005647 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
67. Pachano, T., Haro, E. & Rada-Iglesias, A. Enhancer-gene specificity in development and disease. Development149, dev186536 (2022). 10.1242/dev.186536 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
68. Chopra, V. S., Cande, J., Hong, J.-W. & Levine, M. Stalled Hox promoters as chromosomal boundaries. Genes Dev23, 1505–1509 (2009). 10.1101/gad.1807309 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
69. Vlaming, H., Mimoso, C. A., Field, A. R., Martin, B. J. E. & Adelman, K. Screening thousands of transcribed coding and non-coding regions reveals sequence determinants of RNA polymerase II elongation potential. Nat. Struct. Mol. Biol.29, 613–620 (2022). 10.1038/s41594-022-00785-9 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
70. Viiri, L. E. et al. Extensive reprogramming of the nascent transcriptome during iPSC to hepatocyte differentiation. Sci. Rep.9, 3562 (2019). 10.1038/s41598-019-39215-0 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
71. Gaertner, B. & Zeitlinger, J. RNA polymerase II pausing during development. Development141, 1179–1183 (2014). 10.1242/dev.088492 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
72. Williams, L. H. et al. Pausing of RNA polymerase II regulates mammalian developmental potential through control of signaling networks. Mol. Cell58, 311–322 (2015). 10.1016/j.molcel.2015.02.003 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
73. Ge, X. et al. Outward-oriented sites within clustered CTCF boundaries are key for intra-TAD chromatin interactions and gene regulation. Nat. Commun.14, 8101 (2023). 10.1038/s41467-023-43849-0 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
74. Nanni, L., Ceri, S. & Logie, C. Spatial patterns of CTCF sites define the anatomy of TADs and their boundaries. Genome Biol.21, 197 (2020). 10.1186/s13059-020-02108-x [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
75. Simeone, A. Positioning the isthmic organizer where Otx2 and Gbx2meet. Trends Genet.16, 237–240 (2000). 10.1016/S0168-9525(00)02000-X [Abstract] [CrossRef] [Google Scholar]
76. Tai, C.-I. & Ying, Q.-L. Gbx2, a LIF/Stat3 target, promotes reprogramming to and retention of the pluripotent ground state. J. Cell Sci.126, 1093–1098 (2013). 10.1242/jcs.118273 [Abstract] [CrossRef] [Google Scholar]
77. Anania, C. et al. In vivo dissection of a clustered-CTCF domain boundary reveals developmental principles of regulatory insulation. Nat. Genet.54, 1026–1036 (2022). 10.1038/s41588-022-01117-9 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
78. Huang, H. et al. CTCF mediates dosage- and sequence-context-dependent transcriptional insulation by forming local chromatin domains. Nat. Genet.53, 1064–1074 (2021). 10.1038/s41588-021-00863-6 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
79. Karr, J. P., Ferrie, J. J., Tjian, R. & Darzacq, X. The transcription factor activity gradient (TAG) model: contemplating a contact-independent mechanism for enhancer-promoter communication. Genes Dev.36, 7–16 (2022). 10.1101/gad.349160.121 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
80. Zuin, J. et al. Nonlinear control of transcription through enhancer-promoter interactions. Nature604, 571–577 (2022). 10.1038/s41586-022-04570-y [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
81. Kane, L. et al. Cohesin is required for long-range enhancer action at the Shh locus. Nat. Struct. Mol. Biol.29, 891–897 (2022). 10.1038/s41594-022-00821-8 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
82. Gómez-Marín, C. et al. Evolutionary comparison reveals that diverging CTCF sites are signatures of ancestral topological associating domains borders. Proc. Natl Acad. Sci. USA112, 7542–7547 (2015). 10.1073/pnas.1505463112 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
83. O’Brien, L. L. et al. Transcriptional regulatory control of mammalian nephron progenitors revealed by multi-factor cistromic analysis and genetic studies. PLOS Genet.14, e1007181 (2018). 10.1371/journal.pgen.1007181 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
84. Batut, P. J. et al. Genome organization controls transcriptional dynamics during development. Science375, 566–570 (2022). 10.1126/science.abi7178 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
85. Ngan, C. Y. et al. Chromatin interaction analyses elucidate the roles of PRC2-bound silencers in mouse development. Nat. Genet.52, 264–272 (2020). 10.1038/s41588-020-0581-x [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
86. de Laat, W. & Duboule, D. Topology of mammalian developmental enhancers and their regulatory landscapes. Nature502, 499–506 (2013). 10.1038/nature12753 [Abstract] [CrossRef] [Google Scholar]
87. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell153, 307–319 (2013). 10.1016/j.cell.2013.03.035 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
88. Ringel, A. R. et al. Repression and 3D-restructuring resolves regulatory conflicts in evolutionarily rearranged genomes. Cell185, 3689–3704.e21 (2022). 10.1016/j.cell.2022.09.006 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
89. Roider, H. G., Lenhard, B., Kanhere, A., Haas, S. A. & Vingron, M. CpG-depleted promoters harbor tissue-specific transcription factor binding signals—implications for motif overrepresentation analyses. Nucleic Acids Res.37, 6305–6315 (2009). 10.1093/nar/gkp682 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
90. Dejosez, M. et al. Regulatory architecture of housekeeping genes is driven by promoter assemblies. Cell Rep.42, 112505 (2023). 10.1016/j.celrep.2023.112505 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
91. Kikuta, H. et al. Genomic regulatory blocks encompass multiple neighboring genes and maintain conserved synteny in vertebrates. Genome Res.17, 545–555 (2007). 10.1101/gr.6086307 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
92. Wu, H.-J. et al. Topological isolation of developmental regulators in mammalian genomes. Nat. Commun.12, 4897 (2021). 10.1038/s41467-021-24951-7 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
93. Bateman, J. R. & Johnson, J. E. Altering enhancer–promoter linear distance impacts promoter competition in cis and in trans. Genetics222, iyac098 (2022). 10.1093/genetics/iyac098 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
94. Waymack, R., Gad, M. & Wunderlich, Z. Molecular competition can shape enhancer activity in the Drosophila embryo. iScience24, 103034 (2021). 10.1016/j.isci.2021.103034 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
95. Xiao, J. Y., Hafner, A. & Boettiger, A. N. How subtle changes in 3D structure can create large changes in transcription. eLife10, e64320 (2021). 10.7554/eLife.64320 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
96. Chen, L.-F. et al. Structural elements promote architectural stripe formation and facilitate ultra-long-range gene regulation at a human disease locus. Mol. Cell83, 1446–1461.e6 (2023). 10.1016/j.molcel.2023.03.009 [Abstract] [CrossRef] [Google Scholar]
97. Andrey, G. et al. A switch between topological domains underlies HoxD genes collinearity in mouse limbs. Science340, 1234167 (2013). 10.1126/science.1234167 [Abstract] [CrossRef] [Google Scholar]
98. Hafner, A. et al. Loop stacking organizes genome folding from TADs to chromosomes. Mol. Cell83, 1377–1392.e6 (2023). 10.1016/j.molcel.2023.04.008 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
99. Lupiáñez, D. G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell161, 1012–1025 (2015). 10.1016/j.cell.2015.04.004 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
100. Ewels, P., Magnusson, M., Lundin, S. & Käller, M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics32, 3047–3048 (2016). 10.1093/bioinformatics/btw354 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
101. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics30, 2114–2120 (2014). 10.1093/bioinformatics/btu170 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
102. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods9, 357–359 (2012). 10.1038/nmeth.1923 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
103. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics25, 2078–2079 (2009). 10.1093/bioinformatics/btp352 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
104. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res.44, W160–W165 (2016). 10.1093/nar/gkw257 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
105. Downes, D. J. et al. Capture-C: a modular and flexible approach for high-resolution chromosome conformation capture. Nat. Protoc.17, 445–475 (2022). 10.1038/s41596-021-00651-w [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
106. Buckle, A., Gilbert, N., Marenduzzo, D. & Brackley, C. A. capC-MAP: software for analysis of Capture-C data. Bioinformatics35, 4773–4775 (2019). 10.1093/bioinformatics/btz480 [Abstract] [CrossRef] [Google Scholar]
107. Kent, W. J., Zweig, A. S., Barber, G., Hinrichs, A. S. & Karolchik, D. BigWig and BigBed: enabling browsing of large distributed datasets. Bioinformatics26, 2204–2207 (2010). 10.1093/bioinformatics/btq351 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
108. Feng, J., Liu, T., Qin, B., Zhang, Y. & Liu, X. S. Identifying ChIP-seq enrichment using MACS. Nat. Protoc.7, 1728–1740 (2012). 10.1038/nprot.2012.101 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
109. Hounkpe, B. W., Chenou, F., de Lima, F. & De Paula, E. V. HRT Atlas v1.0 database: redefining human and mouse housekeeping genes and candidate reference transcripts by mining massive RNA-seq datasets. Nucleic Acids Res.49, D947–D955 (2021). 10.1093/nar/gkaa609 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
110. Carbon, S. et al. AmiGO: online access to ontology and annotation data. Bioinformatics25, 288–289 (2009). 10.1093/bioinformatics/btn615 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
111. Wang, Y. et al. The 3D Genome Browser: a web-based browser for visualizing 3D genome organization and long-range chromatin interactions. Genome Biol19, 151 (2018). 10.1186/s13059-018-1519-9 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
112. Hinrichs, A. S. et al. The UCSC Genome Browser Database: update 2006. Nucleic Acids Res.34, D590–D598 (2006). 10.1093/nar/gkj144 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
113. Karolchik, D. et al. The UCSC Table Browser data retrieval tool. Nucleic Acids Res.32, D493–D496 (2004). 10.1093/nar/gkh103 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
114. Liao, Y., Wang, J., Jaehnig, E. J., Shi, Z. & Zhang, B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res.47, W199–W205 (2019). 10.1093/nar/gkz401 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
115. Reiff, S. B. et al. The 4D Nucleome Data Portal as a resource for searching and visualizing curated nucleomics data. Nat. Commun.13, 2365 (2022). 10.1038/s41467-022-29697-4 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
116. Open2C et al.Cooltools: enabling high-resolution Hi-C analysis in Python. PLoS Comput. Biol.20, e1012067 (2024). 10.1371/journal.pcbi.1012067 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
117. Abdennur, N. & Mirny, L. A. Cooler: scalable storage for Hi-C data and other genomically labeled arrays. Bioinformatics36, 311–316 (2020). 10.1093/bioinformatics/btz540 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
118. Dixon, J. R. et al. Chromatin architecture reorganization during stem cell differentiation. Nature518, 331–336 (2015). 10.1038/nature14222 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
119. Hintermann, A. et al. Developmental and evolutionary comparative analysis of a regulatory landscape in mouse and chicken. Development149, dev200594 (2022). 10.1242/dev.200594 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
120. Franke, M. et al. CTCF knockout in zebrafish induces alterations in regulatory landscapes and developmental gene expression. Nat. Commun.12, 5415 (2021). 10.1038/s41467-021-25604-5 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
121. Lee, D. K. Alternatives to P value: confidence interval and effect size. Korean J. Anesthesiol.69, 555–562 (2016). 10.4097/kjae.2016.69.6.555 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
122. Pope, B. D. et al. Topologically associating domains are stable units of replication-timing regulation. Nature515, 402–405 (2014). 10.1038/nature13986 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]

Articles from Nature Communications are provided here courtesy of Nature Publishing Group

Citations & impact 


This article has not been cited yet.

Impact metrics

Jump to Data

Alternative metrics

Altmetric item for https://www.altmetric.com/details/166602374
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/166602374

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

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.

Funding 


Funders who supported this work.

EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 Marie Skłodowska-Curie Actions (H2020 Excellent Science - Marie Skłodowska-Curie Actions) (2)

European Research Council (2)

Ministry of Economy and Competitiveness | Agencia Estatal de Investigación (Spanish Agencia Estatal de Investigación) (2)