Abstract
Recent burst of new technologies that allow for quantitatively delineating chromatin structure has greatly expanded our understanding of how the genome is organized in the three-dimensional (3D) space of the nucleus. It is now clear that the hierarchical organization of the eukaryotic genome critically impacts nuclear activities such as transcription, replication, as well as cellular and developmental events such as cell cycle, cell fate decision and embryonic development. In this review, we discuss new insights into how the structural features of the 3D genome hierarchy are established and maintained, how this hierarchy undergoes dynamic rearrangement during normal development and how its perturbation will lead to human disease, highlighting the accumulating evidence that links the diverse 3D genome architecture components to a multitude of human diseases and the emerging mechanisms by which 3D genome derangement causes disease phenotypes.
Keywords: Chromatin, Chromosome, Hi-C, Three-dimensional (3D) genome architecture, 3D genome organizer, Chromatin structural protein, Topologically associated domain (TAD), Disease
1. Introduction
The genome, the major hereditary materials of the cell, resides in the nucleus, which serves as the cell’s information processing center and controls the various activities of the cell, such as proliferation, homeostasis and division. Early biochemical and microscopy studies have revealed that the nucleus is not geometrically homogenous but rather highly compartmentalized, with the various nuclear activities organized into discrete, functionally-specialized sub-nuclear structures, called nuclear bodies. For example, the nucleolus, which assembles around the rDNA genes, is the largest nuclear body and the primary site of rRNA biogenesis and assembly of ribosomes [1]. Other types of nuclear bodies include Cajal body, Clastosome, Nuclear speckle, Paraspeckle, Nuclear gems, PML body, Histone locus body, and Polycomb body (reviewed in [2, 3]). Moreover, biochemical studies have demonstrated that the two most fundamental nuclear activities, transcription and replication, are also organized in discrete nuclear foci in mammalian nuclei, termed transcription and replication factories, respectively [4, 5]. Meanwhile, the nuclear subcompartments near the nucleoli and nuclear envelope (except the nuclear pores) provide a transcriptionally silent microenvironment for heterochromatic regions to reside, which is critical for maintaining the genome integrity.
For the nearly two meters of genomic DNA to fit into the “tiny” nucleus with a diameter at the scale of several micrometers and to function properly, the genome in every human cell, like all the other eukaryotic genomes, is folded into string-like compact structures, chromatin fibers, whose other essential components are proteins and RNAs [6]. Thus, in addition to the primary (i.e., the DNA double helix) and secondary (i.e., the nucleosomes) structures, the genomic DNA in the nuclear space of eukaryotic cells also possesses a higher-order 3D organization (Fig 1). While it is well established how DNA wraps into nucleosomes, both the underlying mechanisms instructing nucleosomes to fold into chromatin and further to adopt higher-order structures and the molecular details of this process have long remained elusive and debatable. It is only until the past few years, with the significant advance in microscopy-based DNA imaging technologies and the development of high throughput genomic tools for quantitatively measuring chromatin interactions, enormous new insights into chromatin folding and 3D organization have been obtained. For example, recent microscopy studies suggested that chromatin fibers are flexible and disordered chains assembled from both nucleosomes and nucleosome-depleted DNA [7, 8], with a diameter ranging from 5 nm to 24 nm in human cells [8]. It is clear that, despite with some intrinsic stochastic properties, the 3D genome organization is nonrandom and of high functional relevance. It also appears that the spatial arrangement of the genome can be adapted to accomplishing cellular functions other than the genome function per se, such as cell migration, mechanotransduction and vision in nocturnal animals (reviewed in [9]).
Fig 1. Hierarchical organization of the genome.
Genomic DNA in the eukaryotic nucleus possesses multiple levels of organization. The primary structure of the genome here refers to the linear genomic DNA sequences, which harbors the information of DNA modification (e.g., DNA methylation) and genomic distribution of the various types of genes. The secondary structure refers to the nucleosome organization of chromatin. Nucleosomes are considered as the basic unit of chromatin and elicit about 7-fold linear compaction of genomic DNA. This level of structure provides a framework for further assembling the genomic DNA into the chromatin fiber and higher-order structures, as well as a diversity of regulatory mechanisms for genome functions, such as nucleosome positioning, histone modifications and chromatin accessibility. The 3D genome architecture, i.e., the higher-order organization of the genome in the 3D space of the nucleus, comprises multiple levels of topological features, including chromatin loops, sub-megabase-scale self-interacting domains (e.g., TADs), megabase-scale A/B compartments and chromosome territories. Chromatin looping is the fundamental mechanism for building the 3D architecture of chromatin. Promoter-enhancer loops provide a key mechanism for long-range gene regulation. Recent studies have suggested that TADs might be the basic structural and functional units of chromatin. Studies indicate that promoter-enhancer interactions occur much more frequently within TAD than between TADs. Each chromatin fiber is demarcated into transcriptionally active A or inactive B compartments that are defined by large-scale chromatin states. In mammalian cells, each interphase chromosome occupies a distinct nuclear space to form chromosome territories (CT). The radial position of a CT, the relative position between CTs, and the interactions between a CT and the nuclear landmarks are all of functional relevance. The visualization of the results of example assays for each level of structure is shown. The disease relevance of each level of structure is also indicated.
In this review, we briefly introduce our current understanding of the chromatin folding and the spatial genome organizationgained through recent technological developments. We then review emerging evidence linking the disruption of the different components and layers of the 3D genome organization to a range of human diseases, highlighting some of the important questions remaining to be addressed and the potential directions for new technology development in this fast advancing field.
2. Features of the 3D genome organization in the eukaryotic nucleus
Over the past decade, advances in both microscopic and DNA sequencing-based technologies, especially the development and application of the chromosome conformation capture (3C)-derived high throughput genomic methods for mapping chromatin interactions (Box 1), have yielded remarkable new insights into the chromatin folding principles, the organizational features and the structure-functionrelationship of the 3D genome [10–24].
2.1. Hierarchical organization of eukaryotic genomes in the nucleus
High-throughput 3C methods (e.g., Hi-C) have revealed that eukaryotic genomes are nonrandomly organized into a nested hierarchy in the nucleus [25–27]. This hierarchy consists of at least four distinct levels: whole chromosome territories (CTs) [28], large-scale active and repressive compartments (A/B compartments) [29], domains, e.g. topologically associated (TAD) [30, 31], lamina associated (LAD) [32, 33] or nucleolus associate (NAD) [34, 35], and chromatin loops [36]. Early microscopy studies have revealed that individual chromosomes in mammalian cells occupy distinct nuclear space (CTs), with only a limited degree of intermingling between CTs [28, 37], which was later recaptured by Hi-C studies [29]. Individual CTs show preferences for nuclear positioning in mammalian cells, which may correlate with genomic properties. In general, large and gene-poor chromosomes tend to be located near the nuclear periphery, whereas small and gene-rich chromosomes group together near the center of the nucleus [28, 38]. Hi-C studies have also revealed that within individual CTs, chromosomes are partitioned into large compartments at the multi-Mb scale, containing either the active and open (A compartments) or inactive and closed chromatin (B compartments) [29]. The open A compartments consist of high GC-content regions, are gene rich, and are generally highly transcribed. In contrast, B compartments are gene-poor, and less transcriptionally active [29]. However, it remains to be established the extent to which A/B compartments are correlated with the classic, cytogenetically defined euchromatin/heterochromatin. A/B compartments are comprised of TADs, which are considered the basic units of chromosome and genome organization [14, 25, 26]. TADs are self-interacting domains and, seem to be conserved among cell types, tissues, and species; however, the extent of this conservation remains unclear [30, 31]. Chromatin looping is an intrinsic property of chromatin fiber and the fundamental mechanism for building the 3D genome hierarchy [16].
2.2. Dynamics of the 3D genome during cellular and developmental events
Although the 3D genome organization is robust overall, it is characterized by dynamics and can undergo marked changes in the context of different biological conditions. Both the intrinsic randomness, which is attributed to the biophysical properties of chromatin fiber and other related macromolecules in the nucleus, and the constant influence of the DNA-based nuclear activities contribute to the dynamic nature of the 3D genome architecture [16]. The intrinsic randomness likely leads to the cell-to-cell variability of the 3D genome in individual cells. For example, recent super-resolution microscopy and single-cell Hi-C studies have noticed that the formation and the epigenetic states of TADs and chromatin loops within a TAD are highly heterogeneous between individual single cells [39–41]. The nuclear activity-based dynamics of the 3D genome is multifaceted. First, the 3D genome undergoes continuous changes during the cell cycle. It has long been observed in microscopy studies that chromosomes in proliferating mammalian cells are highly condensed in the mitotic phase to facilitate chromosome segregation, but decondensed in interphase to accommodate the diverse nuclear processes. Recent Hi-C studies have revealed that mitotic chromosomes fold into a homogenous linear instead of hierarchy structure that is locus- and cell type-independent, consistent with arrays of consecutive chromatin loops[42, 43]. More recent single-cell Hi-C studies have further revealed that the different levels of the 3D genome organization, A/B compartments, TADs and long-range loops, are governed by distinct cell-cycle dynamics and all undergo continued changes throughout the cell cycle [44]. However, it is noteworthy that similar chromatin domains have been observed in both interphase and mitotic chromosomes by super-resolution live-cell imaging [45]. Second, the 3D chromatin structure can be allelic-specific. The 3D conformation of the inactive and active X chromosomes in both mouse and human female cells is strikingly different, with the inactive X chromosome exhibiting a unique bipartite structure [46–48]. Third, the 3D genome architecture can exhibit distinct features in different tissue/cell types. For example, tight packaging of the sperm genome leads to more long-range chromatin contacts compared to fibroblasts [49, 50], whereas, the 3D genome in mature metaphase II oocytes is similar to that of mitotic cells, which lacks detectable TADs and A/B compartments [50, 51]. It has also been revealed that, compared to proliferating cells, different stages of senescent cells show changes in the frequency of chromatin contacts [52, 53]. Furthermore, comparison of chromatin contact maps in 21 primary human tissues and cell types has uncovered a class of highly tissue-specific genome organizational features termed FIREs, which are local interaction hotspots and correspond to active enhancers [54]. Fourth, global and local rearrangements of the 3D genome occur during cell differentiation, reprogramming, and tissue development. A/B compartment switching, changes in the number and size of TADs, switching of the epigenetic and transcriptional states of TADs, changes in the metaTADs organization, rearrangements of the repressive domains including LADs, NADs and polycomb domains, and rewiring of promoter-enhancer interactions have all been observed during both in vitro and in vivo cell differentiation [33, 55–61], during human brain development [62], and during somatic cell reprograming [63, 64]. Fifth, the 3D genome architecture undergoes striking reprogramming during early mammalian development. Two recent studies of mapping chromatin interactions in mouse gametes and early embryos have revealed several features of the reorganization of the 3D genome during early embryogenesis, including that (i) chromatin exists in a markedly relaxed state after fertilization, showing greatly diminished higher-order structure with very weak TADs and sparse distal interactions compared to those of later stage embryos; (ii) the maternal and paternal chromosomes are spatially separated from each other and display distinct compartmentalization in zygotes, which can be found as late as the 8-cell stage; (iii) establishment of chromatin higher-order architecture is a progressive and multi-level hierarchical process that lasts through preimplantation development; (iv) the establishment of the 3D genome architecture requires DNA replication but is at least partially independent of zygotic genome activation; and (v) chromatin higher-order structures are associated with DNA methylation state, histone modification state and chromatin accessibility [50, 51]. Similar observations have also been obtained at the single-cell level [65, 66]. However, it is noteworthy that, at the single-cell level, TADs and chromatin loops are present at similar strength in zygotic maternal and paternal nuclei, and A/B compartments are notably absent from maternal chromatin, suggesting that compartments and TADs are formed by different mechanisms [65, 66]. And finally, the 3D genome can be reorganized in response to environmental stimuli [67, 68]. It has been shown that hormone treatment can induce gene activity-related structural changes of TADs in human cells [67, 68], whereas, heat shock-induced polycomb-mediated silencing led to widespread rearrangement of 3D genome organization in fly cells [67, 68].
2.3. Roles of genome organizers in the formation and maintenance of the 3D genome
The establishment and maintenance of the 3D genome hierarchy requires participation of proteins and RNAs, in addition to DNA sequence elements, the so called cis determinants (e.g., protein binding sites, housekeeping genes, and macrosatellite repeats). Accumulating evidence has demonstrated the existence of genome organizers that participate in the formation, maintenance, or rearrangement of the 3D genome architecture. These organizers include chromatin architectural proteins such as the CCCTC-binding factor (CTCF)[57], the structural maintenance of chromosomes (SMC) protein complexes (e.g., cohesin and condensin [69–71]) and HP1α [72–74], nuclear matrix proteins (e.g., the lamina proteins [75]), DNA-binding transcription factors [76, 77] (e.g., YY1 [78, 79], SATB1 [80] and SATB2 [81]), chromatin remodeling complexes (e.g., the polycomb complex (PcG) proteins [82] and the NuRD components [41], coactivators (e.g., mediators [83]), and noncoding RNAs such as Xist [84], Firre [85, 86], HOTTIP [87] and ThymoD [88].
Recent studies have underscored the critical role of CTCF and cohesin in the hierarchical chromatin folding, from chromatin looping to the establishment and maintenance of TADs and compartments [25, 57, 89–96]. According to the prominent loop extrusion model, CTCF and SMC proteins, cohesion or condensing, work together, i.e., cohesion or condensin acts as the extruding factor and CTCF as the boundary protein, to create unknotted chromatin loops, which in turn leads to the formation of TADs [97–99]. Indeed, condensin has recently been demonstrated as a mechanochemical motor [100] and the process of DNA loop extrusion by condensin has been visualized in real-time at the single-molecular level [101]. The DNA loop extrusion model is able to explain the diverse observations from Hi-C studies, such as the preferential orientation of CTCF motifs [97, 102–104] and enrichments of CTCF and cohesin at TAD boundaries. It has further been suggested that CTCF, the cohesin release factor Wapl and transcription guide the positioning of cohesin in mammalian genomes [96], and that Wapl and the cohesin loading factor NIPBL together control the extension of chromatin loops and the formation of TADs [95]. However, the direct evidence that clarifies the role of CTCF and cohesin in the 3D genome formation only came from recent loss-of-function studies [92–94]. Acute CTCF depletion in mouse embryonic stem cells using the auxin-inducible degron system revealed that CTCF is absolutely required for CTCF-mediated chromatin looping and TAD formation and functions in a dose-dependent manner [94]. However, it appears that CTCF depletion did not affect the organization of A/B compartments, supporting that TADs and A/B compartments are formed by independent mechanisms [94]. Similar results, i.e., disruption of loops and TADs without destroying A/B compartments, were also observed when cohesin was removed [92, 93], indicating that CTCF and cohesin act together to form loops and TADs but not A/B compartments. The two recent studies, one that removed cohesin from the genome in mouse liver cells by deleting the cohesin loading factor NIPBL [93] and the other that depleted the core cohesin component RAD21 in human colon-cancer cells [92], have also suggested that chromatin state defines cohesin-independent segregation of the genome into fine-scale compartments and that the cohesin-dependent formation of TADs has a role in guiding distant enhancers to their target genes.
In addition to CTCF and cohesin, the roles of other proteins in organizing the 3D genome are emerging. For example, HP1α is well established as an organizer of heterochromatin [105], and recent studies has further suggested that the formation of heterochromatin domain in both fly and mammalian cells might be driven by HP1α-mediated phase separation, i.e., upon DNA binding or phosphorylation, the HP1α protein nucleates into phase-separated droplets through oligomerization, which in turn induces compaction of DNA strands [72–74]. Another well-characterized example are the PcG proteins, which regulate key developmental genes and can guide the 3D genome organization at multiple levels by modifying histones, mediating chromatin looping, and organizing TADs [82]. More recently, the transcription factor YY1 has also emerged as a chromatin structural protein that promotes chromatin looping between promoters and enhancers, a prevalent feature of mammalian gene regulation [78, 79].
Recent studies have identified many RNAs, mainly long noncoding RNAs (lncRNAs) that associate with chromatin in eukaryotic cells [106–110], indicating a structural role of lncRNAs in 3D genome organization. Among them, Xist and Firr are two of the well-characterized lncRNAs that play important roles in organizing the 3D chromatin architecture. Firre has been shown to mediate the colocalization of several genomic regions, located on different chromosomes [85, 86]. The lncRNA Xist guides the formation of the 3D conformation of the inactive X chromosome during X chromosome inactivation [108, 111–113]. Moreover, transcription of the lncRNA ThymoD can guide chromatin folding and compartmentalization to direct promoter-enhancer interactions specifying T cell fate [88]. However, despite these sporadic cases, future studies are required to determine whether and how nuclear lncRNAs play a general role in establishing and maintaining 3D genome architecture.
2.4. The functional relevance of the 3D genome
Existing evidence suggests that the formation of the hierarchical 3D genome is at least partially independent of transcription[50, 114]. However, it is now clear that each level of the 3D genome hierarchy is of functional relevance and that the establishment of the 3D genome organization provides multiple additional regulatory layers to control gene expression (Fig 2), which are complementary to the epigenetic mechanisms mediated by histone modification, DNA methylation or noncoding RNAs [5, 18, 115, 116].
Fig 2. Functional relevance and pathological implications of the various architectural features of the 3D genome.
Cartoons in the left panel show the different topological features for building the 3D genome, including chromatin looping, TAD organization, gene clustering, chromatin compartmentalization and nuclear positioning. These features provide the structural framework for the 3D organization of the various genomic activities. Genome functions related to the 3D organization such as physical interactions between promoters and enhancers during gene transcription, gene looping for coordinating transcription and splicing, coordinated gene expression in transcription factories and the organization and timing of DNA replication in replication foci/factories, are shown in the middle panel. Examples of their pathological consequences are shown in the right panel. More descriptions can be found in the main text. Note, in the cartoon illustrating the replication foci/factories in the bottom of the middle panel, early-replicating domains in the replication factory are indicated in green whereas the late-replicating domains outside are shown in black; In the cartoon showing the long-range effect of SNVs in the top of the right panel, a pathogenic SNV (single-nucleotide variation, the red star) in the enhancer of the gene 1 leads to the silencing of gene 1, and also causes the silencing the two coordinately regulated genes in the vicinity proximity (i.e., in the same transcription factory) via chromatin looping or gene clustering.
Chromatin condensation can regulate the accessibility of transcription factors to DNA, whereas spatial compartmentalization can constrain the availability of nuclear resources (e.g., transcription factors and co-factors). In mammalian cells, transcriptionally silent regions localize near the nuclear envelope and peri-nucleolar space, whereas active regions occupy the space in between [38, 117, 118]. LADs, lamina-associated domains, refer to the regions of the genome that interact with the nuclear lamina at the interior of the nuclear envelope [32, 119, 120]. LADs are transcriptionally repressed chromatin domains and generally enriched in repressive histone modifications [32, 119, 120]. Repositioning of active genes to LADs can result in their repression [111, 121]. Similar to LADs, nucleolus-associated domains (NADs) are also gene poor, and typically characterized by repetitive DNA elements of the centromeric and pericentromeric regions [34, 35].
Emerging evidence suggests that TADs are both structural and functional units that constrain, guide and facilitate enhancer-promoter interactions and coordinate gene regulation [14, 26, 122–124]. Thus, disruption of TAD boundaries can lead to aberrant gene regulation [97, 103, 125, 126]. Also, it has been found that TADs represent replication time domains in mammalian cells [127, 128]. At the fine-scale, chromatin looping can bring distant genomic regions into physical proximity, and chromatin looping-mediated physical contacts between gene promoters and other cis-regulatory elements (e.g., enhancers) are important for gene transcription and for coordinating transcription with RNA splicing [129–132]. The human genome contains many thousands of enhancers that, in any given human cell, are often located distantly from the genes they control [129–132]. Recent genome editing studies demonstrated a causal link between physical promoter-enhancer interactions and gene expression [133, 134]. Furthermore, recent studies showed that YY1-mediated enhancer-promoter interactions are a general mechanism of mammalian gene regulation [78, 79]. TAD formation enables physical co-localization and thereby coordinated gene expression (e.g., an enhancer regulates multiple genes or multiple enhancers regulate one gene within a TAD or a transcription factory), as well as insulation of un-desired enhancer-gene communications. Indeed, it appears that promoter-enhancer interactions occur much more frequently within a TAD than between TADs [122, 123], and each TAD often contains several genes and multiple enhancers, allowing for coordinated gene regulation [14, 26].
3. Biological relevance of the 3D genome organization for human disease
3.1. Implications of the 3D genome hierarchy for human disease
To this end, it appears that chromatin looping, organization into TADs and spatial compartmentalization are three fundamental mechanisms for organizing the genome in 3D and thereby to modulate the DNA-templated nuclear processes, including transcription, splicing, DNA repair and replication. In accordance with their roles in physiological conditions these mechanisms can have potential pathogenic consequences (Fig 2). For example, during recurrent chromosome translocation events, which frequently occur in certain cancer types such as hematologic malignancies and sarcomas, 3D genome-guided spatial proximity strongly influences translocation partner choice [135–140]. Also, coordinated gene transcription within TADs or transcription factories can lead to trans-splicing events that join exons from two different transcripts to produce chimeric mRNAs, a phenomenon underlying several oncogenic fusion transcripts [141, 142]. Moreover, chromatin looping and TAD organization can provide a mechanism to magnify the long-range transcriptional and epigenetic effect of noncoding genetic variants (Fig 2). As disease-risk single nucleotide variants (SNVs) often reside in distal cis-regulatory elements (e.g., enhancers and super-enhancers), they can affect human complex traits or disease phenotypes by inducing changes of the chromatin states at interacting regulatory elements and consequently target gene expression. This demonstrates that the 3D genome organization can guide how SNVs influence distal molecular phenotypes[143–145]. For example, in a recent HiChiP (a method for mapping chromatin interactions mediated by a protein of interest, see Box 1) study, it was found that 684 autoimmune disease–associated intergenic SNVs can influence 2,597 target genes through chromatin interactions, with up to ten target genes for a single SNV [146]. Finally, nuclear compartmentalization also has pathogenic implications, highlighting the importance of nuclear positioning in gene regulation and DNA replication timing [147] (Fig 2). Indeed, the nuclear position of genes or entire chromosomes often differs in disease cells [148–151]. Furthermore, recent studies demonstrated that 3D chromatin architecture is a major influence on both regional mutation rates of SNVs and the landscape of somatic copy-number alterations (SCNAs) in cancer, with SNVs enriched in regions of closed chromatin and insertions and deletions (indels) enriched in open chromatin regions [152–156] (Fig 2).
3.2. Emerging disease mechanisms of 3D genome disruption
Recent studies highlighted a link between the disruption of the 3D genome and human diseases. In human diseases, 3D genome derangements are frequently caused by either deleterious mutations of the 3D genome organizers or the various genetic alterations of the genome, including SNVs, small insertion/deletions (indels), and chromosomal abnormalities (aneuploidy and structural variations (SVs), including insertions, deletions, duplications, translocations and inversions). Although, in most cases, it remains elusive whether a disease-associated 3D genome derangement is the cause or consequence of disease development, it is tempting to postulate that aberrant 3D genome architecture causes dysregulation of genome function (e.g., aberrant gene expression, dysregulation of DNA replication and repair), and thereby leads to disease phenotypes (Fig 3A). For example, abnormal DNA replication caused by derangements in the 3D genome can affect cell cycle and cell division whereas incorrect DNA repair can affect genome integrity and maintenance and thus cause genome instability, both of which can trigger cancer development (Fig 3A).
Fig 3. Disease mechanisms of 3D genome disruption.
A) Schematic illustration of the molecular pathways by which genetic or epigenetic disruption of the 3D genome lead to disease phenotypes; B) cartoons showing that disease-causing noncoding SNVs can disrupt normal gene promoter-enhancer loops (i) or create aberrant promoter-enhancer loops (ii), and thereby leading to aberrant regulation of the disease-relevant genes; C) six examples of genetic or epigenetic mechanisms of TAD disruption that cause enhancer adoption/hijacking and consequently mis-expression of disease-driving genes.
Recent studies indicated that there are at least two mechanisms by which disruption of 3D genome architecture causes altered expression of disease-relevant genes and thereby disease phenotypes (Fig 3). In the first mechanism, disease-associated regulatory SNVs impair normal or create pathogenic promoter-enhancer interactions and thereby lead to aberrant gene expression and disease phenotypes (Fig 3B). A large body of studies have demonstrated that this is an important and widely used mechanism for regulatory noncoding variants to play a role in many human diseases. For example, point mutations in the ZRS, a distal enhancer that tightly regulates the sonic hedgehog (SHH) expression in the developing limb, cause ectopic chromatin interactions between ZRS and the SHH promoter and thereby lead to SHH misexpressions in several congenital limb malformation disorders [157]. In another example, a recent study found that many coronary artery disease (CAD)-risk noncoding SNVs identified by genome-wide association studies (GWAS) exerted their disease effect either by disrupting or by strengthening enhancer–promoter interactions [146]. Similar analyses of chromatin interactions at disease-associated loci identified many novel candidate genes for a diversity of complex diseases, including inflammatory bowel disease [158], chronic kidney disease (CKD) [159], atherosclerotic disease [160], autoimmune disease [146, 161], and prostate cancer [162, 163]. Moreover, a recent study developed a computational approach that is based on chromatin interactions for predicting cancer-driving mutations in noncoding regions [164]. Together, these studies demonstrated that disease-linked regulatory SNVs modulate target gene expression by regulating chromatin interactions genes and regulatory elements.
In the second disease mechanism, disruption of TADs leads to inappropriate communications between enhancers and genes that are normally insulated from each other by TAD organization and thereby allows distal enhancers to ectopically activate disease-relevant genes. This mechanism, called enhancer adoption or enhancer hijacking [165–168], underscores the importance of TAD organization for genomic integrity and disease. As discussed above, TADs are thought to function as regulatory units of gene expression by constraining promoter-enhancer interactions. Hence, it is not surprising that TAD disruption can cause rewiring of promoter-enhancer interactions and subsequent gene misregulation. Indeed, recent studies have identified a variety of pathogenic events that lead to enhancer adoption/hijacking via TAD disruption [169–171] (Fig. 3C). For example, since TADs are demarcated by boundary regions that are characterized by the binding of architectural proteins (e.g., CTCF and cohesin), genetic disruption or epigenetic inactivation of the architectural protein binding sites (i.e., CTCF binding sites) in a TAD boundary can lead to fusion of the two flanking TADs, affecting the encompassing genes [58, 172–174]. Similarly, deletion of an entire TAD boundary will also lead to TAD fusion or disruption [172, 175]. Moreover, complex genomic rearrangements such as inversions, deletions, duplications and translocations has the potential to cause TAD breakage and fusion or even formation of new TADs [166, 167, 172, 176–178].
Several pioneering studies have identified TAD disruption-mediated enhancer adoption and gene misregulation as a novel mechanism for structure variation (SV)-caused congenital developmental diseases (Table 1). In the first such studies, the authors linked the phenotypes of 922 deletion cases recorded in the DECIPHER database to monogenic diseases that are associated with genes within or adjacent to the deletions using a bioinformatic approach. They found up to 11.8% of the deletions could result in TAD disruption and lead to enhancer adoption [166]. In a more recent study with 273 subjects harboring balanced chromosomal abnormalities (BCA) associated with a spectrum of human congenital anomalies, the authors found that 7.3% of the BCAs recurrently disrupted TADs encompassing known disease-relevant genes. They identified disruptions of the TAD organization that cause long-range regulatory perturbation of several disease-driving genes such as the MEF2C gene in the patients with 5q14.3 microdeletion syndrome [179] (Table 1). However, the first direct evidence demonstrating TAD disruption as a disease mechanism in human developmental disorders came from a recent mechanistic study of the pathogenicity of SVs in the limb malformations [172]. In this elegant study, the authors first identified the genomic rearrangements (deletions, inversions and duplications) associated with three types of human limb malformations, and found that these rearrangements altered the TAD organization in the WNT6/IHH/EPHA4/PAX3 locus. The authors then re-engineered the same rearrangements in mice by using CRISPR/CAS9 genome editing, and found that these pathogenic structural changes led to ectopic long-range promoter-enhancer communications and thereby disease-causing gene misexpression both in the mutant mouse limb tissue and patient-derived fibroblasts. The authors further found that this rewiring of enhancer-promoter interactions happened only if the boundary elements (e.g., CTCF binding sites) were disrupted, highlighting the importance of the TAD integrity [172]. Furthermore, a recent study from the same research group revealed that genomic duplications cause disease phenotypes of Cook syndrome by leading to the formation of a new TAD (neo-TAD) in the Sox9 locus, which in turn leads to ectopic interaction between the Kcnj2 gene and the Sox9 enhancers and thereby misexpression of kcnj2 [176]. TAD disruption-caused enhancer adoption is also one of the mechanisms underlying the pathogenesis of lamin B1-caused autosomal dominant adult-onset demyelinating leukodystrophy (ADLD)[175]. Together, these studies demonstrate the functional relevance of TAD disruption in determining Mendelian phenotypes of developmental disorders.
Table 1.
Examples of TAD disruption-mediated enhancer adoption/hijacking in human diseases
| Disease type | Disease | Disease- relevant gene |
Genetic /epigenetic alteration |
OMIM ID | Refs. |
|---|---|---|---|---|---|
| Congenital developmental disorders | Type A1 Brachydactyly | PAX3 | Deletion | 112500 | [172] |
| F-syndrome | WNT6 | Inversion, duplication | 102510 | [172] | |
| Cooks syndrome | KCNJ2 | Duplication | 106995 | [176] | |
| Rett syndrome | FOXG1 | Translocation | 613454 | [179] | |
| Glass syndrome | SATB2 | Translocation | 612313 | [179] | |
| 5q14.3 microdeletion syndrome | MEF2C | Deletion | 613443 | [179] | |
| Autosomal-dominant adultonset demyelinating leukodystrophy (ADLD) | LMNB1 | Deletion | 169500 | [175] | |
| Mesomelic dysplasia | ID4 | Deletion | 605274 | [170] | |
| Liebenberg syndrome | PITX1 | Deletion, duplication | 186550 | [170] | |
| Cancer | Glioma | PDGFRA | Hypermethylation of CTCF binding site | - | [174] |
| Colorectal cancer | - | SNVs in CTCF/cohesion binding site | - | [173] | |
| T cell acute lymphoblastic leukemia (T-ALL) | TAL1, LMO2 | Deletion of CTCF/cohesion binding sites | - | [181] | |
| Neuroblastoma | TERT | Diverse SVs | - | [185, 186] | |
| Lung cancer | IRS4 | Deletion | - | [178] | |
| Colorectal cancer | IGF2 | Tandem duplications | - | [178] | |
| Medulloblastoma | GFI1, GFI1B | Diverse SVs | - | [167] | |
| AML with inv(3)/t(3;3) | EVI1 | Inversion | - | [177] |
Similar to enhancer adoption in developmental disorders, recent studies have suggested TAD disruption-mediated enhancer hijacking as an important mechanism of oncogene activation in cancers. For example, a recent study found that gain-of-function IDH mutations promote gliomagenesis by disrupting TAD organization and therefore causing ectopic chromatin interactions between the oncogene PDGFRA and a distal constitutive enhancer that induce PDGFRA deregulation [174]. The authors further demonstrated that the inactivation of the TAD boundary is caused by compromised CTCF binding to the boundary due to DNA hyper-methylation at the CTCF binding sites resulted from IDH mutation [174]. Several lines of evidence have suggested that TAD disruption-caused oncogene activation might be a prevalent mechanism for tumorigenesis. First, it has been demonstrated that CTCF and cohesin binding and CTCF site orientation play a critical role in TAD formation and maintenance, whereas recent studies have found that CTCF/cohesin-binding sites are frequently mutated across numerous cancer types [180], such as colorectal cancer[173] and leukaemia [181]. Second, recurrent mutations in many epigenetic regulators frequently occur as cancer-driving events across a wide range of cancer types [182]. The resulting gain- or loss-of function of these epigenetic regulators can cause epigenetic alterations that in turn may lead to epigenetic inactivation of TAD boundaries in a way similar to how IDH mutations lead to TAD disruption in gliomas [174]. Third, as a hallmark of cancer, oncogenic alterations such as SVs frequently occur in cancer genomes with the potential to disrupt TAD organization and normal promoter-enhancer interactions. Indeed, two recent Hi-C studies, which mapped the 3D genome organization in prostate cancer and multiple myeloma cell lines, respectively, found that copy number variation (CNV) breakpoints significantly overlap with TAD boundaries [183, 184]. These studies also suggested that CNVs might help in the formation of cancer-specific TADs enriched in regulatory elements such as enhancers and promoters, and associated with aberrant gene expression [183, 184]. Moreover, a recent study of pan-cancer analysis of somatic CNVs in 7,416 cancer genomes across 26 tumor types found that TAD boundary disruption caused by recurrent deletions or tandem duplications led to the activation of the two oncogenes IRS4 and IGF2 through enhancer hijacking [178]. And fourth, oncogene activation by TAD disruption-mediated enhancer hijacking has been observed in numerous cancer types, including neuroblastoma [185, 186], medulloblastoma [167], glioma [174], colorectal cancer [178], leukemia [177, 181], sarcoma, uterine leiomyoma and squamous cancers [178]. With more investigations into the 3D cancer genomes forthcoming, one can expect more such examples emerging. Compared to oncogene activation by TAD disruption, much less is known about the pathogenic repression of tumor suppressor genes in relation to the 3D cancer genome. It will be interesting to see whether tumor suppressors can be silenced through aberrant long-range chromatin interactions caused by TAD boundary disruption.
In conclusion, a growing number of studies demonstrated that genetic and epigenetic alterations can cause disease phenotypes by altering the two fundamental layers of the 3D genome hierarchy, chromatin loops and TADs. Additionally, other types of epigenetic alterations with the potential to affect other components of the 3D genome hierarchy, e.g., compartments and CTs, have also been observed in human diseases, as exemplified by pathogenic nuclear repositioning of entire chromosomes or individual genes. For instance, movement of the X chromosome and the Bdnf gene, which encodes the major neurotrophin, in seizure foci of the human cortex is associated with epilepsy [187, 188]. Moreover, as discussed above, such radial repositioning of chromosomes and gene loci also occurs across cancer types [148, 149]. However, it must be pointed out that all the examples discussed here are correlative, and whether nuclear repositioning is causative for human disease remains to be examined.
4. Genome architectural proteins and human diseases
Chromatin architectural proteins act in concert with each other and other genome organizers including transcription factors and ncRNAs to play critical roles in the formation and maintenance of the 3D genome organization, and thus are pivotal in human development, tissue regeneration and disease. Indeed, an increasing number of genes encoding architectural proteins have been linked to a myriad of human diseases, including developmental disorders, neurodegenerative disorders, psychiatric disorders and accelerated aging disorders, as well as cancers (Table 2). For example, mutations and heterozygous deletions of the CTCF gene are implicated in autosomal dominant mental retardation 21 (MRD21) [189, 190] and cancers [91]. Recurrent somatic mutations in the genes that encode the proteins constituting the cohesin complex have also been identified in many tumors such as myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) [191]. In another example, mutations in the gene encoding SATB2, a chromatin organizer that tethers genomic regions to the nuclear matrix and is essential for craniofacial development, cause the Glass syndrome characterized by intellectual disability and various dysmorphic facial features (Table 1). Moreover, mutations in the gene MED12 cause the FG syndrome and Fryns-Lujan syndrome (Table 2). However, among all of the structural protein-associated human diseases, it is the two groups of diseases, cohesinopathies and laminopathies that attract much interest.
Table 2.
Examples of chromatin architectural protein defects-caused human disorders
| Architectural protein |
Disease | Inheritance format | OMIM ID |
|---|---|---|---|
| CTCF | autosomal dominant mental retardation 21 (MRD21) | Autosomal dominant | 615502 |
| MED12 | FG syndrome | X-linked recessive | 305450 |
| MED12 | Fryns-Lujan syndrome | X-linked recessive | 309520 |
| NIPBL | CdLS1 | Autosomal dominant | 122470 |
| SMC1A | CdLS2 | X-linked dominant | 300590 |
| SMC3 | CdLS3 | Autosomal dominant | 610759 |
| RAD21 | CdLS4 | Autosomal dominant | 606462 |
| ESCO2 | Roberts syndrome | Autosomal recessive | 268300 |
| DDX11 | Warsaw Breakage Syndrome | Autosomal recessive | 613398 |
| Lamin A | Hutchinson–Gilford progeria syndrome (HGPS) | Autosomal dominant | 176670 |
| RECQL2 | Werner syndrome | Autosomal recessive | 277700 |
| Lamin A/C | Emery–Dreifuss muscular dystrophy 2 (EDMD2) | Autosomal dominant | 181350 |
| Lamin A/C | Emery–Dreifuss muscular dystrophy 3 (EDMD3) | Autosomal recessive | 616516 |
| Lamin A/C | limb girdle muscular dystrophy type 1B (LGMD1B) | Autosomal dominant | 159001 |
| Lamin A/C | dilated cardiomyopathy type 1A (DCM1A) | Autosomal dominant | 115200 |
| Lamin A/C | Charcot-Marie-Tooth type 2B1 (CMT2B1) | Autosomal recessive | 605588 |
| Lamin A/C | Dunnigan-type familial partial lipodystrophy (FPLD) | Autosomal dominant | 151660 |
| LBR | Greenberg dysplasia | Autosomal recessive | 215140 |
| LBR | Pelger–Huet anomaly (PHA) | Autosomal dominant | 169400 |
Genetic mapping and DNA sequencing studies have revealed that mutations in the genes encoding the cohesin complex proteins and their regulators are responsible for cohesinopathies, a group of developmental and intellectual impairment diseases that include the Cornelia de Lange Syndrome (CdLS), Roberts syndrome (RBS), and Warsaw Breakage Syndrome (WABS)[192–194]. CdLS is mainly caused by mutations in the cohesin-loading factor NIPBL (>60%) as well as mutations in the cohesin subunits Smc1A, Smc3 and Rad21 (~5%). RBS, an autosomal recessive disorder, arises from mutations in the gene encoding cohesin acetyltransferase ESCO2, whereas WABS is caused by defective DDX11, a DNA helicase essential for chromatid cohesion. The cohesinopathies are characterized by a diversity of overlapping phenotypes including limb defects, craniofacial deformities, growth retardation, intellectual disability, cardiac malformations, and microcephaly, which is consistent with the multifaceted roles of cohesin proteins in mitotic chromosome condensation, sister chromatid cohesion, gene regulation, DNA repair, 3D genome organization, cell cycle, apoptosis, and tissue development. However, the precise molecular mechanisms underlying these syndromes remain to be elucidated.
Laminopathies are a wide spectrum of rare disorders arisen from mutations in genes encoding the intermediate filament nuclear lamins and lamin B receptor (LBR), which belong to the more generic nuclear envelopathies, diseases associated with defects of the nuclear envelope (reviewed in [195, 196]). The nuclear lamina, composed of three types of filamentous protein (lamin A, B and C), are essential for the nuclear structure and the 3D genome organization by providing structural support to the nucleus and interacting with the genome. For example, about 40% of the human genome is organized into LADs, which range from 40 kb to 30 Mb. The nuclear lamin proteins therefore play diverse roles in many nuclear processes and cellular pathways, such as nuclear positioning, heterochromatin organization, gene transcription, nuclear morphology, metabolism, mechanosensation, and cell locomotion. Hence, similar to cohesinopathies, laminopathies are also multisystem monogenic diseases, i.e., caused by a single genetic defect in a single gene but showing phenotypes in multiple tissues or organs. However, laminopathies exhibit a distinct feature, i.e., different mutations in the same gene often result in different disorders. For example, mutations in LMNA encoding the A/C-type lamins cause a large group of disorders ranging from premature ageing syndromes (Hutchinson–Gilford progeria syndrome (HGPS) and Werner syndrome) to myopathies (autosomal forms of Emery–Dreifuss muscular dystrophy (EDMD), limb girdle muscular dystrophy type 1B (LGMD1B) and dilated cardiomyopathy type 1A (DCM1A)), neuropathies (e.g., Charcot-Marie-Tooth type 2B1 (CMT2B1)) and Lipodystrophies (e.g., Dunnigan-type familial partial lipodystrophy (FPLD)) (Table 2). Also, different mutations in the gene encoding LBR cause at least two disorders, Greenberg dysplasia, a lethal and recessive chondrodystrophy, and the Pelger–Huet anomaly (PHA) (Table 2). PHA is an autosomal dominant disorder, characterized by hypolobulated neutrophil nuclei with coarse chromatin. Interestingly, the nuclear morphology changes in neutrophils resembling PHA that are acquired rather than congenital have also been described in a diversity of disease conditions, such as MDS, vitamin B12 and folate deficiency, and multiple myeloma, indicating that aberrant nuclear structure might be a common mechanism underlying these diseases.
Taken together, these congenital disorders caused by defective structural proteins showcase the critical roles of genome organizers in the various aspects of human development, tissue regeneration and aging processes. However, there is no clear understanding yet regarding how the 3D genome organization is disrupted as a result of the altered genome organizer in almost any of these diseases. Thus, whether or how disruption of the 3D genome causes the disease phenotypes remains to be elucidated. Future studies that comprehensively map the derangements in the 3D genome in these diseases and dissect the link between the derangements and disease phenotypes will likely uncover new disease mechanisms and identify novel therapeutic targets.
5. Conclusions and future prospects
The synthesis of a wide array of technologies ranging from the still rapidly developing DNA imaging technologies to the fast-growing high-throughput genomic tools for mapping chromatin interactions, the CRISPR/CAS genome engineering technology and the computational data analysis and modelling has greatly facilitated our understanding of the physical organization of the genome in the 3D nuclear space. It is becoming clear that the eukaryotic genome is organized as a nested hierarchy that comprises multiple levels of topological features including chromatin loops, TADs, A/B compartments and CTs. It appears that the hierarchical strata of the 3D genome provide multiple regulatory layers for gene regulation and other genome functions and undergo programmed spatiotemporal reorganization during animal development. Moreover, defects in the 3D genome at each layer of the organization accompanied by various genetic and epigenetic alterations have been observed in human disease. Although many aspects of the 3D genome architecture in human disease and their underlying mechanisms begin to emerge, future studies have to overcome several challenges to better understand the role of the spatial genome organization in disease.
Our understanding of the 3D genome and its regulatory roles in the various nuclear processes is still far from complete. There are many fundamental questions remaining with respect to how the genome is organized and functions. For example, how is TAD, suggested as the functional unit of gene regulation, related to the formation and nuclear localization of a transcription factory during transcription? In other words, does a transcription factory represent the physical existence of an active TAD or several active TADs? How is the specificity of a chromatin loop between a gene and its enhancer within a TAD achieved? How is a compartment (A or B) related to the nuclear positioning of its encompassing genomic loci? What factor(s) determines the A/B compartment state of a gene or a genomic locus in a given cell? To answer these important questions, new mapping technologies for spatial resolution of the 3D genome architecture are needed. Indeed, developing new mapping technologies is one of the main focuses of the 4D Nucleome Project [197]. Moreover, genome editing studies aimed at determining the functional consequences of perturbing the various structural features in a variety of biological contexts are also required. Such experiments will offer deeper mechanistic insights into the principles of the 3D genome organization and how perturbation of the different organizational features will affect nuclear activities, critical to understand the molecular mechanisms of how disruption of the 3D genome will cause disease.
Our understanding of how the spatiotemporal organization of the genome orchestrates normal tissue development is also very limited. Although the dynamic rearrangement of the 3D genome has recently been described during the early mouse embryonic development, during in vitro differentiation of mouse and human ESCs, and during somatic cell reprogramming, mechanistic insights into how the spatiotemporal organization of the nucleus orchestrates normal tissue development remains elusive. For example, what are the driving forces that govern the 3D genome rearrangement during the various tissue-specific cell differentiation processes? Whether and how does the 3D genome impact the cell fate decision during these differentiation processes? Whether and how does the rearrangement of the 3D genome play a role in the precise spatiotemporal regulation of gene expression during tissue development? More specifically, for example, how does the 3D genome influence hematopoiesis, the process that produces all types of blood cells? It is well documented that the various blood precursors undergo dramatic nuclear morphological changes during blood cell maturation. For example, throughout terminal erythropoiesis, erythroblasts undergo gradual chromatin condensation with decreased cell and nucleus size, which ultimately leads to enucleation in red blood cells, whereas granulopoiesis is characterized by dramatic and specific changes in nuclear shape, with the nucleus segmented into lobes in mature neutrophils. It is unclear how the nuclear morphological change is linked to the 3D genome rearrangement and transcriptional re-wiring during blood differentiation. To address such questions, perturbation studies in the context of tissue development using the genome editing technologies will again be required. These experiments will not only shed light on the structure-function relationships of the genome in the context of normal tissue development, but also be critical to eventually understand the link between aberrant 3D genome architecture and human disease phenotypes. For example, aberrant nuclear morphologies are hallmarks for the diagnosis of many hematologic disorders, such as MDS. Thus, defining the functional relevance of the nuclear morphology-associated 3D genome rearrangement during normal and dysplastic hematopoiesis is of great importance both biologically and clinically.
There is a lack of reference maps of the 3D genome in human primary normal or diseased tissue cells. To this end, except a few studies[54, 62], the vast majority of whole-genome chromatin interaction maps have been generated in cell lines. In particularly, mapping of 3D cancer genomes is by far exclusively limited to cancer cell lines [183, 184]. Hence, a straightforward next step is to construct high-resolution reference maps of the 3D genome in human disease cells and their corresponding normal tissue cells. However, the cellular and genetic heterogeneity of primary disease samples, especially the heterogeneity within primary tumor samples, presents a daunting challenge to perform cell population-based high-throughput mapping studies. Thus, the recently developed single-cell Hi-C methods (Box 1) offer the promise of overcoming this technically challenge. Moreover, cell-to-cell variability of chromosome conformation in individual cells also requires surveying relatively large numbers of single cells before achieving statistically meaningful observations in single-cell assays. However, the current implements of the single-cell Hi-C methods have limitations either in throughput (i.e., the number of single cells being surveyed in a single experiment) or in resolution (i.e., the recovered distinct chromatin interactions per single cell). The scHi-C [44, 198] and snHi-C [65] enable to detect up to two millions of chromatin contacts per cell but can only survey a limited number of cells per assay, whereas sciHi-C [199] enables to survey thousands of cells per assay but with relative low resolution. Thus, it is in great need to develop high-throughput and high-resolution single-cell Hi-C methods for dissecting the aberrant 3D genome architecture in the heterogeneous human primary disease samples.
Acknowledgments
This work was supported by the UW Bridge Fund (ZD), ASH Bridge Grant (ZD), and the NIH Common Fund U54DK107979.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Competing financial interests
The authors declare no competing interests.
References
- 1.Matheson TD, Kaufman PD. Grabbing the genome by the NADs. Chromosoma. 2015 doi: 10.1007/s00412-015-0527-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Dundr M, Misteli T. Biogenesis of nuclear bodies. Cold Spring Harbor perspectives in biology. 2010;2(12):a000711. doi: 10.1101/cshperspect.a000711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mao YS, Zhang B, Spector DL. Biogenesis and function of nuclear bodies. Trends in genetics : TIG. 2011;27(8):295–306. doi: 10.1016/j.tig.2011.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cook PR. A model for all genomes: the role of transcription factories. Journal of molecular biology. 2010;395(1):1–10. doi: 10.1016/j.jmb.2009.10.031. [DOI] [PubMed] [Google Scholar]
- 5.Misteli T, Soutoglou E. The emerging role of nuclear architecture in DNA repair and genome maintenance. Nature reviews. Molecular cell biology. 2009;10(4):243–54. doi: 10.1038/nrm2651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Felsenfeld G, Groudine M. Controlling the double helix. Nature. 2003;421(6921):448–53. doi: 10.1038/nature01411. [DOI] [PubMed] [Google Scholar]
- 7.Ricci MA, Manzo C, Garcia-Parajo MF, Lakadamyali M, Cosma MP. Chromatin fibers are formed by heterogeneous groups of nucleosomes in vivo. Cell. 2015;160(6):1145–58. doi: 10.1016/j.cell.2015.01.054. [DOI] [PubMed] [Google Scholar]
- 8.Ou HD, Phan S, Deerinck TJ, Thor A, Ellisman MH, O'Shea CC. ChromEMT: Visualizing 3D chromatin structure and compaction in interphase and mitotic cells. Science. 2017;357(6349) doi: 10.1126/science.aag0025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bustin M, Misteli T. Nongenetic functions of the genome. Science. 2016;352(6286):aad6933. doi: 10.1126/science.aad6933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bickmore WA. The spatial organization of the human genome. Annual review of genomics and human genetics. 2013;14:67–84. doi: 10.1146/annurev-genom-091212-153515. [DOI] [PubMed] [Google Scholar]
- 11.Cavalli G, Misteli T. Functional implications of genome topology. Nature structural & molecular biology. 2013;20(3):290–9. doi: 10.1038/nsmb.2474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cremer T, Cremer M, Hubner B, Strickfaden H, Smeets D, Popken J, Sterr M, Markaki Y, Rippe K, Cremer C. The 4D nucleome: Evidence for a dynamic nuclear landscape based on co-aligned active and inactive nuclear compartments. FEBS letters. 2015;589(20 Pt A):2931–43. doi: 10.1016/j.febslet.2015.05.037. [DOI] [PubMed] [Google Scholar]
- 13.de Wit E, de Laat W. A decade of 3C technologies: insights into nuclear organization. Genes & development. 2012;26(1):11–24. doi: 10.1101/gad.179804.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Dekker J, Heard E. Structural and functional diversity of Topologically Associating Domains. FEBS letters. 2015;589(20 Pt A):2877–84. doi: 10.1016/j.febslet.2015.08.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Denker A, de Laat W. The second decade of 3C technologies: detailed insights into nuclear organization. Genes & development. 2016;30(12):1357–82. doi: 10.1101/gad.281964.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Duan Z, Blau CA. The genome in space and time: does form always follow function? How does the spatial and temporal organization of a eukaryotic genome reflect and influence its functions? Bioessays. 2012;34(9):800–10. doi: 10.1002/bies.201200034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fraser J, Williamson I, Bickmore WA, Dostie J. An Overview of Genome Organization and How We Got There: from FISH to Hi-C. Microbiology and molecular biology reviews : MMBR. 2015;79(3):347–72. doi: 10.1128/MMBR.00006-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gorkin DU, Leung D, Ren B. The 3D genome in transcriptional regulation and pluripotency. Cell stem cell. 2014;14(6):762–75. doi: 10.1016/j.stem.2014.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Misteli T. Beyond the sequence: cellular organization of genome function. Cell. 2007;128(4):787–800. doi: 10.1016/j.cell.2007.01.028. [DOI] [PubMed] [Google Scholar]
- 20.Pombo A, Dillon N. Three-dimensional genome architecture: players and mechanisms. Nature reviews. Molecular cell biology. 2015;16(4):245–57. doi: 10.1038/nrm3965. [DOI] [PubMed] [Google Scholar]
- 21.Rajapakse I, Groudine M. On emerging nuclear order. The Journal of cell biology. 2011;192(5):711–21. doi: 10.1083/jcb.201010129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ramani V, Shendure J, Duan Z. Understanding Spatial Genome Organization: Methods and Insights. Genomics, proteomics & bioinformatics. 2016;14(1):7–20. doi: 10.1016/j.gpb.2016.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Risca VI, Greenleaf WJ. Unraveling the 3D genome: genomics tools for multiscale exploration. Trends in genetics : TIG. 2015 doi: 10.1016/j.tig.2015.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tjong H, Li W, Kalhor R, Dai C, Hao S, Gong K, Zhou Y, Li H, Zhou XJ, Le Gros MA, Larabell CA, Chen L, Alber F. Population-based 3D genome structure analysis reveals driving forces in spatial genome organization. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(12):E1663–72. doi: 10.1073/pnas.1512577113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bouwman BA, de Laat W. Getting the genome in shape: the formation of loops. domains and compartments, Genome biology. 2015;16:154. doi: 10.1186/s13059-015-0730-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dixon JR, Gorkin DU, Ren B. Chromatin Domains: The Unit of Chromosome Organization. Molecular cell. 2016;62(5):668–80. doi: 10.1016/j.molcel.2016.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gibcus JH, Dekker J. The hierarchy of the 3D genome. Molecular cell. 2013;49(5):773–82. doi: 10.1016/j.molcel.2013.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cremer T, Cremer M. Chromosome territories. Cold Spring Harbor perspectives in biology. 2010;2(3):a003889. doi: 10.1101/cshperspect.a003889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A, Amit I, Lajoie BR, Sabo PJ, Dorschner MO, Sandstrom R, Bernstein B, Bender MA, Groudine M, Gnirke A, Stamatoyannopoulos J, Mirny LA, Lander ES, Dekker J. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science. 2009;326(5950):289–93. doi: 10.1126/science.1181369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, Hu M, Liu JS, Ren B. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature. 2012 doi: 10.1038/nature11082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Nora EP, Lajoie BR, Schulz EG, Giorgetti L, Okamoto I, Servant N, Piolot T, van Berkum NL, Meisig J, Sedat J, Gribnau J, Barillot E, Bluthgen N, Dekker J, Heard E. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature. 2012 doi: 10.1038/nature11049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Guelen L, Pagie L, Brasset E, Meuleman W, Faza MB, Talhout W, Eussen BH, de Klein A, Wessels L, de Laat W, van Steensel B. Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions. Nature. 2008;453(7197):948–51. doi: 10.1038/nature06947. [DOI] [PubMed] [Google Scholar]
- 33.Peric-Hupkes D, Meuleman W, Pagie L, Bruggeman SW, Solovei I, Brugman W, Graf S, Flicek P, Kerkhoven RM, van Lohuizen M, Reinders M, Wessels L, van Steensel B. Molecular maps of the reorganization of genome-nuclear lamina interactions during differentiation. Molecular cell. 2010;38(4):603–13. doi: 10.1016/j.molcel.2010.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Nemeth A, Conesa A, Santoyo-Lopez J, Medina I, Montaner D, Peterfia B, Solovei I, Cremer T, Dopazo J, Langst G. Initial genomics of the human nucleolus. PLoS genetics. 2010;6(3):e1000889. doi: 10.1371/journal.pgen.1000889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.van Koningsbruggen S, Gierlinski M, Schofield P, Martin D, Barton GJ, Ariyurek Y, den Dunnen JT, Lamond AI. High-resolution whole-genome sequencing reveals that specific chromatin domains from most human chromosomes associate with nucleoli. Molecular biology of the cell. 2010;21(21):3735–48. doi: 10.1091/mbc.E10-06-0508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, Sanborn AL, Machol I, Omer AD, Lander ES, Aiden EL. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell. 2014;159(7):1665–80. doi: 10.1016/j.cell.2014.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gondor A, Ohlsson R. Chromosome crosstalk in three dimensions. Nature. 2009;461(7261):212–7. doi: 10.1038/nature08453. [DOI] [PubMed] [Google Scholar]
- 38.Takizawa T, Meaburn KJ, Misteli T. The meaning of gene positioning. Cell. 2008;135(1):9–13. doi: 10.1016/j.cell.2008.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang S, Su JH, Beliveau BJ, Bintu B, Moffitt JR, Wu CT, Zhuang X. Spatial organization of chromatin domains and compartments in single chromosomes. Science. 2016;353(6299):598–602. doi: 10.1126/science.aaf8084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Boettiger AN, Bintu B, Moffitt JR, Wang S, Beliveau BJ, Fudenberg G, Imakaev M, Mirny LA, Wu CT, Zhuang X. Super-resolution imaging reveals distinct chromatin folding for different epigenetic states. Nature. 2016;529(7586):418–22. doi: 10.1038/nature16496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Stevens TJ, Lando D, Basu S, Atkinson LP, Cao Y, Lee SF, Leeb M, Wohlfahrt KJ, Boucher W, O'Shaughnessy-Kirwan A, Cramard J, Faure AJ, Ralser M, Blanco E, Morey L, Sanso M, Palayret MGS, Lehner B, Di Croce L, Wutz A, Hendrich B, Klenerman D, Laue ED. 3D structures of individual mammalian genomes studied by single-cell Hi-C. Nature. 2017;544(7648):59–64. doi: 10.1038/nature21429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Naumova N, Imakaev M, Fudenberg G, Zhan Y, Lajoie BR, Mirny LA, Dekker J. Organization of the mitotic chromosome. Science. 2013;342(6161):948–53. doi: 10.1126/science.1236083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gibcus JH, Samejima K, Goloborodko A, Samejima I, Naumova N, Nuebler J, Kanemaki MT, Xie L, Paulson JR, Earnshaw WC, Mirny LA, Dekker J. A pathway for mitotic chromosome formation. Science. 2018 doi: 10.1126/science.aao6135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nagano T, Lubling Y, Varnai C, Dudley C, Leung W, Baran Y, Mendelson Cohen N, Wingett S, Fraser P, Tanay A. Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature. 2017;547(7661):61–67. doi: 10.1038/nature23001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Nozaki T, Imai R, Tanbo M, Nagashima R, Tamura S, Tani T, Joti Y, Tomita M, Hibino K, Kanemaki MT, Wendt KS, Okada Y, Nagai T, Maeshima K. Dynamic Organization of Chromatin Domains Revealed by Super-Resolution Live-Cell Imaging. Molecular cell. 2017;67(2):282–293. e7. doi: 10.1016/j.molcel.2017.06.018. [DOI] [PubMed] [Google Scholar]
- 46.Deng X, Ma W, Ramani V, Hill A, Yang F, Ay F, Berletch JB, Blau CA, Shendure J, Duan Z, Noble WS, Disteche CM. Bipartite structure of the inactive mouse X chromosome. Genome biology. 2015;16:152. doi: 10.1186/s13059-015-0728-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Giorgetti L, Lajoie BR, Carter AC, Attia M, Zhan Y, Xu J, Chen CJ, Kaplan N, Chang HY, Heard E, Dekker J. Structural organization of the inactive X chromosome in the mouse. Nature. 2016;535(7613):575–9. doi: 10.1038/nature18589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Darrow EM, Huntley MH, Dudchenko O, Stamenova EK, Durand NC, Sun Z, Huang SC, Sanborn AL, Machol I, Shamim M, Seberg AP, Lander ES, Chadwick BP, Aiden EL. Deletion of DXZ4 on the human inactive X chromosome alters higher-order genome architecture. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(31):E4504–12. doi: 10.1073/pnas.1609643113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Battulin N, Fishman VS, Mazur AM, Pomaznoy M, Khabarova AA, Afonnikov DA, Prokhortchouk EB, Serov OL. Comparison of the three-dimensional organization of sperm and fibroblast genomes using the Hi-C approach. Genome biology. 2015;16:77. doi: 10.1186/s13059-015-0642-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ke Y, Xu Y, Chen X, Feng S, Liu Z, Sun Y, Yao X, Li F, Zhu W, Gao L, Chen H, Du Z, Xie W, Xu X, Huang X, Liu J. 3D Chromatin Structures of Mature Gametes and Structural Reprogramming during Mammalian Embryogenesis. Cell. 2017;170(2):367–381. e20. doi: 10.1016/j.cell.2017.06.029. [DOI] [PubMed] [Google Scholar]
- 51.Du Z, Zheng H, Huang B, Ma R, Wu J, Zhang X, He J, Xiang Y, Wang Q, Li Y, Ma J, Zhang X, Zhang K, Wang Y, Zhang MQ, Gao J, Dixon JR, Wang X, Zeng J, Xie W. Allelic reprogramming of 3D chromatin architecture during early mammalian development. Nature. 2017;547(7662):232–235. doi: 10.1038/nature23263. [DOI] [PubMed] [Google Scholar]
- 52.Chandra T, Ewels PA, Schoenfelder S, Furlan-Magaril M, Wingett SW, Kirschner K, Thuret JY, Andrews S, Fraser P, Reik W. Global reorganization of the nuclear landscape in senescent cells. Cell reports. 2015;10(4):471–83. doi: 10.1016/j.celrep.2014.12.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Criscione SW, De Cecco M, Siranosian B, Zhang Y, Kreiling JA, Sedivy JM, Neretti N. Reorganization of chromosome architecture in replicative cellular senescence. Science advances. 2016;2(2):e1500882. doi: 10.1126/sciadv.1500882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Schmitt AD, Hu M, Jung I, Xu Z, Qiu Y, Tan CL, Li Y, Lin S, Lin Y, Barr CL, Ren B. A Compendium of Chromatin Contact Maps Reveals Spatially Active Regions in the Human Genome. Cell reports. 2016;17(8):2042–2059. doi: 10.1016/j.celrep.2016.10.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Dixon JR, Jung I, Selvaraj S, Shen Y, Antosiewicz-Bourget JE, Lee AY, Ye Z, Kim A, Rajagopal N, Xie W, Diao Y, Liang J, Zhao H, Lobanenkov VV, Ecker JR, Thomson JA, Ren B. Chromatin architecture reorganization during stem cell differentiation. Nature. 2015;518(7539):331–6. doi: 10.1038/nature14222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Fraser J, Ferrai C, Chiariello AM, Schueler M, Rito T, Laudanno G, Barbieri M, Moore BL, Kraemer DC, Aitken S, Xie SQ, Morris KJ, Itoh M, Kawaji H, Jaeger I, Hayashizaki Y, Carninci P, Forrest AR, Consortium F, Semple CA, Dostie J, Pombo A, Nicodemi M. Hierarchical folding and reorganization of chromosomes are linked to transcriptional changes in cellular differentiation. Molecular systems biology. 2015;11(12):852. doi: 10.15252/msb.20156492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Phillips-Cremins JE, Sauria ME, Sanyal A, Gerasimova TI, Lajoie BR, Bell JS, Ong CT, Hookway TA, Guo C, Sun Y, Bland MJ, Wagstaff W, Dalton S, McDevitt TC, Sen R, Dekker J, Taylor J, Corces VG. Architectural protein subclasses shape 3D organization of genomes during lineage commitment. Cell. 2013;153(6):1281–95. doi: 10.1016/j.cell.2013.04.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ji X, Dadon DB, Powell BE, Fan ZP, Borges-Rivera D, Shachar S, Weintraub AS, Hnisz D, Pegoraro G, Lee TI, Misteli T, Jaenisch R, Young RA. 3D Chromosome Regulatory Landscape of Human Pluripotent Cells. Cell stem cell. 2016;18(2):262–75. doi: 10.1016/j.stem.2015.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Rubin AJ, Barajas BC, Furlan-Magaril M, Lopez-Pajares V, Mumbach MR, Howard I, Kim DS, Boxer LD, Cairns J, Spivakov M, Wingett SW, Shi M, Zhao Z, Greenleaf WJ, Kundaje A, Snyder M, Chang HY, Fraser P, Khavari PA. Lineage-specific dynamic and preestablished enhancer-promoter contacts cooperate in terminal differentiation. Nature genetics. 2017;49(10):1522–1528. doi: 10.1038/ng.3935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bonev B, Mendelson Cohen N, Szabo Q, Fritsch L, Papadopoulos GL, Lubling Y, Xu X, Lv X, Hugnot JP, Tanay A, Cavalli G. Multiscale 3D Genome Rewiring during Mouse Neural Development. Cell. 2017;171(3):557–572. e24. doi: 10.1016/j.cell.2017.09.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Javierre BM, Burren OS, Wilder SP, Kreuzhuber R, Hill SM, Sewitz S, Cairns J, Wingett SW, Varnai C, Thiecke MJ, Burden F, Farrow S, Cutler AJ, Rehnstrom K, Downes K, Grassi L, Kostadima M, Freire-Pritchett P, Wang F, Consortium B, Stunnenberg HG, Todd JA, Zerbino DR, Stegle O, Ouwehand WH, Frontini M, Wallace C, Spivakov M, Fraser P. Lineage-Specific Genome Architecture Links Enhancers and Non-coding Disease Variants to Target Gene Promoters. Cell. 2016;167(5):1369–1384. e19. doi: 10.1016/j.cell.2016.09.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Won H, de la Torre-Ubieta L, Stein JL, Parikshak NN, Huang J, Opland CK, Gandal MJ, Sutton GJ, Hormozdiari F, Lu D, Lee C, Eskin E, Voineagu I, Ernst J, Geschwind DH. Chromosome conformation elucidates regulatory relationships in developing human brain. Nature. 2016;538(7626):523–527. doi: 10.1038/nature19847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Beagan JA, Gilgenast TG, Kim J, Plona Z, Norton HK, Hu G, Hsu SC, Shields EJ, Lyu X, Apostolou E, Hochedlinger K, Corces VG, Dekker J, Phillips-Cremins JE. Local Genome Topology Can Exhibit an Incompletely Rewired 3D-Folding State during Somatic Cell Reprogramming. Cell stem cell. 2016;18(5):611–24. doi: 10.1016/j.stem.2016.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Krijger PH, Di Stefano B, de Wit E, Limone F, van Oevelen C, de Laat W, Graf T. Cell-of-Origin-Specific 3D Genome Structure Acquired during Somatic Cell Reprogramming. Cell stem cell. 2016;18(5):597–610. doi: 10.1016/j.stem.2016.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Flyamer IM, Gassler J, Imakaev M, Brandao HB, Ulianov SV, Abdennur N, Razin SV, Mirny LA, Tachibana-Konwalski K. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature. 2017;544(7648):110–114. doi: 10.1038/nature21711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Gassler J, Brandao HB, Imakaev M, Flyamer IM, Ladstatter S, Bickmore WA, Peters JM, Mirny LA, Tachibana K. A mechanism of cohesin-dependent loop extrusion organizes zygotic genome architecture. The EMBO journal. 2017;36(24):3600–3618. doi: 10.15252/embj.201798083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Le Dily F, Bau D, Pohl A, Vicent GP, Serra F, Soronellas D, Castellano G, Wright RH, Ballare C, Filion G, Marti-Renom MA, Beato M. Distinct structural transitions of chromatin topological domains correlate with coordinated hormone-induced gene regulation. Genes & development. 2014;28(19):2151–62. doi: 10.1101/gad.241422.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Li L, Lyu X, Hou C, Takenaka N, Nguyen HQ, Ong CT, Cubenas-Potts C, Hu M, Lei EP, Bosco G, Qin ZS, Corces VG. Widespread rearrangement of 3D chromatin organization underlies polycomb-mediated stress-induced silencing. Molecular cell. 2015;58(2):216–31. doi: 10.1016/j.molcel.2015.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lioy VS, Cournac A, Marbouty M, Duigou S, Mozziconacci J, Espeli O, Boccard F, Koszul R. Multiscale Structuring of the E. coli Chromosome by Nucleoid-Associated and Condensin Proteins. Cell. 2018 doi: 10.1016/j.cell.2017.12.027. [DOI] [PubMed] [Google Scholar]
- 70.Murayama Y, Samora CP, Kurokawa Y, Iwasaki H, Uhlmann F. Establishment of DNA-DNA Interactions by the Cohesin Ring. Cell. 2018;172(3):465–477. e15. doi: 10.1016/j.cell.2017.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hanssen LLP, Kassouf MT, Oudelaar AM, Biggs D, Preece C, Downes DJ, Gosden M, Sharpe JA, Sloane-Stanley JA, Hughes JR, Davies B, Higgs DR. Tissue-specific CTCF-cohesin-mediated chromatin architecture delimits enhancer interactions and function in vivo. Nature cell biology. 2017;19(8):952–961. doi: 10.1038/ncb3573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Strom AR, Emelyanov AV, Mir M, Fyodorov DV, Darzacq X, Karpen GH. Phase separation drives heterochromatin domain formation. Nature. 2017;547(7662):241–245. doi: 10.1038/nature22989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Larson AG, Elnatan D, Keenen MM, Trnka MJ, Johnston JB, Burlingame AL, Agard DA, Redding S, Narlikar GJ. Liquid droplet formation by HP1alpha suggests a role for phase separation in heterochromatin. Nature. 2017;547(7662):236–240. doi: 10.1038/nature22822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Machida S, Takizawa Y, Ishimaru M, Sugita Y, Sekine S, Nakayama JI, Wolf M, Kurumizaka H. Structural Basis of Heterochromatin Formation by Human HP1. Molecular cell. 2018 doi: 10.1016/j.molcel.2017.12.011. [DOI] [PubMed] [Google Scholar]
- 75.Dechat T, Pfleghaar K, Sengupta K, Shimi T, Shumaker DK, Solimando L, Goldman RD. Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin. Genes & development. 2008;22(7):832–53. doi: 10.1101/gad.1652708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.de Wit E, Bouwman BA, Zhu Y, Klous P, Splinter E, Verstegen MJ, Krijger PH, Festuccia N, Nora EP, Welling M, Heard E, Geijsen N, Poot RA, Chambers I, de Laat W. The pluripotent genome in three dimensions is shaped around pluripotency factors. Nature. 2013;501(7466):227–31. doi: 10.1038/nature12420. [DOI] [PubMed] [Google Scholar]
- 77.Stadhouders R, Vidal E, Serra F, Di Stefano B, Le Dily F, Quilez J, Gomez A, Collombet S, Berenguer C, Cuartero Y, Hecht J, Filion GJ, Beato M, Marti-Renom MA, Graf T. Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming. Nature genetics. 2018 doi: 10.1038/s41588-017-0030-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Beagan JA, Duong MT, Titus KR, Zhou L, Cao Z, Ma J, Lachanski CV, Gillis DR, Phillips-Cremins JE. YY1 and CTCF orchestrate a 3D chromatin looping switch during early neural lineage commitment. Genome research. 2017;27(7):1139–1152. doi: 10.1101/gr.215160.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Weintraub AS, Li CH, Zamudio AV, Sigova AA, Hannett NM, Day DS, Abraham BJ, Cohen MA, Nabet B, Buckley DL, Guo YE, Hnisz D, Jaenisch R, Bradner JE, Gray NS, Young RA. YY1 Is a Structural Regulator of Enhancer-Promoter Loops. Cell. 2017;171(7):1573–1588. e28. doi: 10.1016/j.cell.2017.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Cai S, Lee CC, Kohwi-Shigematsu T. SATB1 packages densely looped, transcriptionally active chromatin for coordinated expression of cytokine genes. Nature genetics. 2006;38(11):1278–88. doi: 10.1038/ng1913. [DOI] [PubMed] [Google Scholar]
- 81.Dobreva G, Dambacher J, Grosschedl R. SUMO modification of a novel MAR-binding protein, SATB2, modulates immunoglobulin mu gene expression. Genes & development. 2003;17(24):3048–61. doi: 10.1101/gad.1153003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Entrevan M, Schuettengruber B, Cavalli G. Regulation of Genome Architecture and Function by Polycomb Proteins. Trends in cell biology. 2016;26(7):511–25. doi: 10.1016/j.tcb.2016.04.009. [DOI] [PubMed] [Google Scholar]
- 83.Kagey MH, Newman JJ, Bilodeau S, Zhan Y, Orlando DA, van Berkum NL, Ebmeier CC, Goossens J, Rahl PB, Levine SS, Taatjes DJ, Dekker J, Young RA. Mediator and cohesin connect gene expression and chromatin architecture. Nature. 2010;467(7314):430–5. doi: 10.1038/nature09380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lee JT. Epigenetic regulation by long noncoding RNAs. Science. 2012;338(6113):1435–9. doi: 10.1126/science.1231776. [DOI] [PubMed] [Google Scholar]
- 85.Hacisuleyman E, Goff LA, Trapnell C, Williams A, Henao-Mejia J, Sun L, McClanahan P, Hendrickson DG, Sauvageau M, Kelley DR, Morse M, Engreitz J, Lander ES, Guttman M, Lodish HF, Flavell R, Raj A, Rinn JL. Topological organization of multichromosomal regions by the long intergenic noncoding RNA Firre. Nature structural & molecular biology. 2014;21(2):198–206. doi: 10.1038/nsmb.2764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Yang F, Deng X, Ma W, Berletch JB, Rabaia N, Wei G, Moore JM, Filippova GN, Xu J, Liu Y, Noble WS, Shendure J, Disteche CM. The lncRNA Firre anchors the inactive X chromosome to the nucleolus by binding CTCF and maintains H3K27me3 methylation. Genome biology. 2015;16:52. doi: 10.1186/s13059-015-0618-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, Lajoie BR, Protacio A, Flynn RA, Gupta RA, Wysocka J, Lei M, Dekker J, Helms JA, Chang HY. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature. 2011;472(7341):120–4. doi: 10.1038/nature09819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Isoda T, Moore AJ, He Z, Chandra V, Aida M, Denholtz M, Piet van Hamburg J, Fisch KM, Chang AN, Fahl SP, Wiest DL, Murre C. Non-coding Transcription Instructs Chromatin Folding and Compartmentalization to Dictate Enhancer-Promoter Communication and T Cell Fate. Cell. 2017;171(1):103–119. e18. doi: 10.1016/j.cell.2017.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Phillips JE, Corces VG. CTCF: master weaver of the genome. Cell. 2009;137(7):1194–211. doi: 10.1016/j.cell.2009.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Bonev B, Cavalli G. Organization and function of the 3D genome. Nature reviews. Genetics. 2016;17(11):661–678. doi: 10.1038/nrg.2016.112. [DOI] [PubMed] [Google Scholar]
- 91.Norton HK, Phillips-Cremins JE. Crossed wires: 3D genome misfolding in human disease. The Journal of cell biology. 2017;216(11):3441–3452. doi: 10.1083/jcb.201611001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Rao SSP, Huang SC, Glenn St Hilaire B, Engreitz JM, Perez EM, Kieffer-Kwon KR, Sanborn AL, Johnstone SE, Bascom GD, Bochkov ID, Huang X, Shamim MS, Shin J, Turner D, Ye Z, Omer AD, Robinson JT, Schlick T, Bernstein BE, Casellas R, Lander ES, Aiden EL. Cohesin Loss Eliminates All Loop Domains. Cell. 2017;171(2):305–320. e24. doi: 10.1016/j.cell.2017.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Schwarzer W, Abdennur N, Goloborodko A, Pekowska A, Fudenberg G, Loe-Mie Y, Fonseca NA, Huber W, C HH, Mirny L, Spitz F. Two independent modes of chromatin organization revealed by cohesin removal. Nature. 2017;551(7678):51–56. doi: 10.1038/nature24281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Nora EP, Goloborodko A, Valton AL, Gibcus JH, Uebersohn A, Abdennur N, Dekker J, Mirny LA, Bruneau BG. Targeted Degradation of CTCF Decouples Local Insulation of Chromosome Domains from Genomic Compartmentalization. Cell. 2017;169(5):930–944. e22. doi: 10.1016/j.cell.2017.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Haarhuis JHI, van der Weide RH, Blomen VA, Yanez-Cuna JO, Amendola M, van Ruiten MS, Krijger PHL, Teunissen H, Medema RH, van Steensel B, Brummelkamp TR, de Wit E, Rowland BD. The Cohesin Release Factor WAPL Restricts Chromatin Loop Extension. Cell. 2017;169(4):693–707. e14. doi: 10.1016/j.cell.2017.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Busslinger GA, Stocsits RR, van der Lelij P, Axelsson E, Tedeschi A, Galjart N, Peters JM. Cohesin is positioned in mammalian genomes by transcription, CTCF and Wapl. Nature. 2017;544(7651):503–507. doi: 10.1038/nature22063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Sanborn AL, Rao SS, Huang SC, Durand NC, Huntley MH, Jewett AI, Bochkov ID, Chinnappan D, Cutkosky A, Li J, Geeting KP, Gnirke A, Melnikov A, McKenna D, Stamenova EK, Lander ES, Aiden EL. Chromatin extrusion explains key features of loop and domain formation in wild-type and engineered genomes. Proc Natl Acad Sci U S A. 2015;112(47):E6456–65. doi: 10.1073/pnas.1518552112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Alipour E, Marko JF. Self-organization of domain structures by DNA-loop-extruding enzymes. Nucleic acids research. 2012;40(22):11202–12. doi: 10.1093/nar/gks925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Fudenberg G, Imakaev M, Lu C, Goloborodko A, Abdennur N, Mirny LA. Formation of Chromosomal Domains by Loop Extrusion. Cell reports. 2016;15(9):2038–49. doi: 10.1016/j.celrep.2016.04.085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Terakawa T, Bisht S, Eeftens JM, Dekker C, Haering CH, Greene EC. The condensin complex is a mechanochemical motor that translocates along DNA. Science. 2017;358(6363):672–676. doi: 10.1126/science.aan6516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ganji M, Shaltiel IA, Bisht S, Kim E, Kalichava A, Haering CH, Dekker C. Real-time imaging of DNA loop extrusion by condensin. Science. 2018 doi: 10.1126/science.aar7831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.de Wit E, Vos ES, Holwerda SJ, Valdes-Quezada C, Verstegen MJ, Teunissen H, Splinter E, Wijchers PJ, Krijger PH, de Laat W. CTCF Binding Polarity Determines Chromatin Looping. Molecular cell. 2015;60(4):676–84. doi: 10.1016/j.molcel.2015.09.023. [DOI] [PubMed] [Google Scholar]
- 103.Guo Y, Xu Q, Canzio D, Shou J, Li J, Gorkin DU, Jung I, Wu H, Zhai Y, Tang Y, Lu Y, Wu Y, Jia Z, Li W, Zhang MQ, Ren B, Krainer AR, Maniatis T, Wu Q. CRISPR Inversion of CTCF Sites Alters Genome Topology and Enhancer/Promoter Function. Cell. 2015;162(4):900–10. doi: 10.1016/j.cell.2015.07.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Tang Z, Luo OJ, Li X, Zheng M, Zhu JJ, Szalaj P, Trzaskoma P, Magalska A, Wlodarczyk J, Ruszczycki B, Michalski P, Piecuch E, Wang P, Wang D, Tian SZ, Penrad-Mobayed M, Sachs LM, Ruan X, Wei CL, Liu ET, Wilczynski GM, Plewczynski D, Li G, Ruan Y. CTCF-Mediated Human 3D Genome Architecture Reveals Chromatin Topology for Transcription. Cell. 2015;163(7):1611–27. doi: 10.1016/j.cell.2015.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Vermaak D, Malik HS. Multiple roles for heterochromatin protein 1 genes in Drosophila. Annual review of genetics. 2009;43:467–92. doi: 10.1146/annurev-genet-102108-134802. [DOI] [PubMed] [Google Scholar]
- 106.Chu C, Qu K, Zhong FL, Artandi SE, Chang HY. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Molecular cell. 2011;44(4):667–78. doi: 10.1016/j.molcel.2011.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Simon MD, Wang CI, Kharchenko PV, West JA, Chapman BA, Alekseyenko AA, Borowsky ML, Kuroda MI, Kingston RE. The genomic binding sites of a noncoding RNA. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(51):20497–502. doi: 10.1073/pnas.1113536108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Engreitz JM, Pandya-Jones A, McDonel P, Shishkin A, Sirokman K, Surka C, Kadri S, Xing J, Goren A, Lander ES, Plath K, Guttman M. The Xist lncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science. 2013;341(6147):1237973. doi: 10.1126/science.1237973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Sridhar B, Rivas-Astroza M, Nguyen TC, Chen W, Yan Z, Cao X, Hebert L, Zhong S. Systematic Mapping of RNA-Chromatin Interactions In Vivo. Current biology : CB. 2017;27(4):602–609. doi: 10.1016/j.cub.2017.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Li X, Zhou B, Chen L, Gou LT, Li H, Fu XD. GRID-seq reveals the global RNA-chromatin interactome. Nature biotechnology. 2017;35(10):940–950. doi: 10.1038/nbt.3968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Chen CK, Blanco M, Jackson C, Aznauryan E, Ollikainen N, Surka C, Chow A, Cerase A, McDonel P, Guttman M. Xist recruits the X chromosome to the nuclear lamina to enable chromosome-wide silencing. Science. 2016 doi: 10.1126/science.aae0047. [DOI] [PubMed] [Google Scholar]
- 112.Splinter E, de Wit E, Nora EP, Klous P, van de Werken HJ, Zhu Y, Kaaij LJ, van Ijcken W, Gribnau J, Heard E, de Laat W. The inactive X chromosome adopts a unique three-dimensional conformation that is dependent on Xist RNA. Genes & development. 2011;25(13):1371–83. doi: 10.1101/gad.633311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Jegu T, Aeby E, Lee JT. The X chromosome in space. Nature reviews. Genetics. 2017;18(6):377–389. doi: 10.1038/nrg.2017.17. [DOI] [PubMed] [Google Scholar]
- 114.Hug CB, Grimaldi AG, Kruse K, Vaquerizas JM. Chromatin Architecture Emerges during Zygotic Genome Activation Independent of Transcription. Cell. 2017;169(2):216–228. e19. doi: 10.1016/j.cell.2017.03.024. [DOI] [PubMed] [Google Scholar]
- 115.Chakalova L, Fraser P. Organization of transcription. Cold Spring Harbor perspectives in biology. 2010;2(9):a000729. doi: 10.1101/cshperspect.a000729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Sexton T, Yaffe E, Kenigsberg E, Bantignies F, Leblanc B, Hoichman M, Parrinello H, Tanay A, Cavalli G. Three-dimensional folding and functional organization principles of the Drosophila genome. Cell. 2012;148(3):458–72. doi: 10.1016/j.cell.2012.01.010. [DOI] [PubMed] [Google Scholar]
- 117.Zimmer C, Fabre E. Principles of chromosomal organization: lessons from yeast. The Journal of cell biology. 2011;192(5):723–33. doi: 10.1083/jcb.201010058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Wijchers PJ, Krijger PH, Geeven G, Zhu Y, Denker A, Verstegen MJ, Valdes-Quezada C, Vermeulen C, Janssen M, Teunissen H, Anink-Groenen LC, Verschure PJ, de Laat W. Cause and Consequence of Tethering a SubTAD to Different Nuclear Compartments. Molecular cell. 2016;61(3):461–73. doi: 10.1016/j.molcel.2016.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kind J, Pagie L, de Vries SS, Nahidiazar L, Dey SS, Bienko M, Zhan Y, Lajoie B, de Graaf CA, Amendola M, Fudenberg G, Imakaev M, Mirny LA, Jalink K, Dekker J, van Oudenaarden A, van Steensel B. Genome-wide maps of nuclear lamina interactions in single human cells. Cell. 2015;163(1):134–47. doi: 10.1016/j.cell.2015.08.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kind J, Pagie L, Ortabozkoyun H, Boyle S, de Vries SS, Janssen H, Amendola M, Nolen LD, Bickmore WA, van Steensel B. Single-cell dynamics of genome-nuclear lamina interactions. Cell. 2013;153(1):178–92. doi: 10.1016/j.cell.2013.02.028. [DOI] [PubMed] [Google Scholar]
- 121.Robson MI, de Las Heras JI, Czapiewski R, Le Thanh P, Booth DG, Kelly DA, Webb S, Kerr AR, Schirmer EC. Tissue-Specific Gene Repositioning by Muscle Nuclear Membrane Proteins Enhances Repression of Critical Developmental Genes during Myogenesis. Molecular cell. 2016;62(6):834–47. doi: 10.1016/j.molcel.2016.04.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Jin F, Li Y, Dixon JR, Selvaraj S, Ye Z, Lee AY, Yen CA, Schmitt AD, Espinoza CA, Ren B. A high-resolution map of the three-dimensional chromatin interactome in human cells. Nature. 2013;503(7475):290–4. doi: 10.1038/nature12644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Ma W, Ay F, Lee C, Gulsoy G, Deng X, Cook S, Hesson J, Cavanaugh C, Ware CB, Krumm A, Shendure J, Blau CA, Disteche CM, Noble WS, Duan Z. Fine-scale chromatin interaction maps reveal the cis-regulatory landscape of human lincRNA genes. Nature methods. 2015;12(1):71–8. doi: 10.1038/nmeth.3205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Symmons O, Pan L, Remeseiro S, Aktas T, Klein F, Huber W, Spitz F. The Shh Topological Domain Facilitates the Action of Remote Enhancers by Reducing the Effects of Genomic Distances. Developmental cell. 2016;39(5):529–543. doi: 10.1016/j.devcel.2016.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Flavahan WA, Drier Y, Liau BB, Gillespie SM, Venteicher AS, Stemmer-Rachamimov AO, Suva ML, Bernstein BE. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature. 2015 doi: 10.1038/nature16490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lupianez DG, Kraft K, Heinrich V, Krawitz P, Brancati F, Klopocki E, Horn D, Kayserili H, Opitz JM, Laxova R, Santos-Simarro F, Gilbert-Dussardier B, Wittler L, Borschiwer M, Haas SA, Osterwalder M, Franke M, Timmermann B, Hecht J, Spielmann M, Visel A, Mundlos S. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell. 2015;161(5):1012–25. doi: 10.1016/j.cell.2015.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Dileep V, Ay F, Sima J, Vera DL, Noble WS, Gilbert DM. Topologically associating domains and their long-range contacts are established during early G1 coincident with the establishment of the replication-timing program. Genome research. 2015;25(8):1104–13. doi: 10.1101/gr.183699.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Pope BD, Ryba T, Dileep V, Yue F, Wu W, Denas O, Vera DL, Wang Y, Hansen RS, Canfield TK, Thurman RE, Cheng Y, Gulsoy G, Dennis JH, Snyder MP, Stamatoyannopoulos JA, Taylor J, Hardison RC, Kahveci T, Ren B, Gilbert DM. Topologically associating domains are stable units of replication-timing regulation. Nature. 2014;515(7527):402–5. doi: 10.1038/nature13986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kadauke S, Blobel GA. Chromatin loops in gene regulation. Biochimica et biophysica acta. 2009;1789(1):17–25. doi: 10.1016/j.bbagrm.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Pennacchio LA, Bickmore W, Dean A, Nobrega MA, Bejerano G. Enhancers: five essential questions. Nature reviews. Genetics. 2013;14(4):288–95. doi: 10.1038/nrg3458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Bulger M, Groudine M. Functional and mechanistic diversity of distal transcription enhancers. Cell. 2011;144(3):327–39. doi: 10.1016/j.cell.2011.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Dekker J, Misteli T. Long-Range Chromatin Interactions. Cold Spring Harbor perspectives in biology. 2015;7(10) doi: 10.1101/cshperspect.a019356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Deng W, Lee J, Wang H, Miller J, Reik A, Gregory PD, Dean A, Blobel GA. Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell. 2012;149(6):1233–44. doi: 10.1016/j.cell.2012.03.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Deng W, Rupon JW, Krivega I, Breda L, Motta I, Jahn KS, Reik A, Gregory PD, Rivella S, Dean A, Blobel GA. Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell. 2014;158(4):849–60. doi: 10.1016/j.cell.2014.05.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Engreitz JM, Agarwala V, Mirny LA. Three-dimensional genome architecture influences partner selection for chromosomal translocations in human disease. PloS one. 2012;7(9):e44196. doi: 10.1371/journal.pone.0044196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Mani RS, Tomlins SA, Callahan K, Ghosh A, Nyati MK, Varambally S, Palanisamy N, Chinnaiyan AM. Induced chromosomal proximity and gene fusions in prostate cancer. Science. 2009;326(5957):1230. doi: 10.1126/science.1178124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nikiforova MN, Stringer JR, Blough R, Medvedovic M, Fagin JA, Nikiforov YE. Proximity of chromosomal loci that participate in radiation-induced rearrangements in human cells. Science. 2000;290(5489):138–41. doi: 10.1126/science.290.5489.138. [DOI] [PubMed] [Google Scholar]
- 138.Roix JJ, McQueen PG, Munson PJ, Parada LA, Misteli T. Spatial proximity of translocation-prone gene loci in human lymphomas. Nat Genet. 2003;34(3):287–91. doi: 10.1038/ng1177. [DOI] [PubMed] [Google Scholar]
- 139.Zhang Y, McCord RP, Ho YJ, Lajoie BR, Hildebrand DG, Simon AC, Becker MS, Alt FW, Dekker J. Spatial organization of the mouse genome and its role in recurrent chromosomal translocations. Cell. 2012;148(5):908–21. doi: 10.1016/j.cell.2012.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hakim O, Resch W, Yamane A, Klein I, Kieffer-Kwon KR, Jankovic M, Oliveira T, Bothmer A, Voss TC, Ansarah-Sobrinho C, Mathe E, Liang G, Cobell J, Nakahashi H, Robbiani DF, Nussenzweig A, Hager GL, Nussenzweig MC, Casellas R. DNA damage defines sites of recurrent chromosomal translocations in B lymphocytes. Nature. 2012;484(7392):69–74. doi: 10.1038/nature10909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Li H, Wang J, Mor G, Sklar J. A neoplastic gene fusion mimics trans-splicing of RNAs in normal human cells. Science. 2008;321(5894):1357–61. doi: 10.1126/science.1156725. [DOI] [PubMed] [Google Scholar]
- 142.Rickman DS, Pflueger D, Moss B, VanDoren VE, Chen CX, de la Taille A, Kuefer R, Tewari AK, Setlur SR, Demichelis F, Rubin MA. SLC45A3-ELK4 is a novel and frequent erythroblast transformation-specific fusion transcript in prostate cancer. Cancer research. 2009;69(7):2734–8. doi: 10.1158/0008-5472.CAN-08-4926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.McVicker G, van de Geijn B, Degner JF, Cain CE, Banovich NE, Raj A, Lewellen N, Myrthil M, Gilad Y, Pritchard JK. Identification of genetic variants that affect histone modifications in human cells. Science. 2013;342(6159):747–9. doi: 10.1126/science.1242429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Waszak SM, Delaneau O, Gschwind AR, Kilpinen H, Raghav SK, Witwicki RM, Orioli A, Wiederkehr M, Panousis NI, Yurovsky A, Romano-Palumbo L, Planchon A, Bielser D, Padioleau I, Udin G, Thurnheer S, Hacker D, Hernandez N, Reymond A, Deplancke B, Dermitzakis ET. Population Variation and Genetic Control of Modular Chromatin Architecture in Humans. Cell. 2015;162(5):1039–50. doi: 10.1016/j.cell.2015.08.001. [DOI] [PubMed] [Google Scholar]
- 145.Grubert F, Zaugg JB, Kasowski M, Ursu O, Spacek DV, Martin AR, Greenside P, Srivas R, Phanstiel DH, Pekowska A, Heidari N, Euskirchen G, Huber W, Pritchard JK, Bustamante CD, Steinmetz LM, Kundaje A, Snyder M. Genetic Control of Chromatin States in Humans Involves Local and Distal Chromosomal Interactions. Cell. 2015;162(5):1051–65. doi: 10.1016/j.cell.2015.07.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Mumbach MR, Satpathy AT, Boyle EA, Dai C, Gowen BG, Cho SW, Nguyen ML, Rubin AJ, Granja JM, Kazane KR, Wei Y, Nguyen T, Greenside PG, Corces MR, Tycko J, Simeonov DR, Suliman N, Li R, Xu J, Flynn RA, Kundaje A, Khavari PA, Marson A, Corn JE, Quertermous T, Greenleaf WJ, Chang HY. Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements. Nature genetics. 2017;49(11):1602–1612. doi: 10.1038/ng.3963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Shachar S, Misteli T. Causes and consequences of nuclear gene positioning. Journal of cell science. 2017;130(9):1501–1508. doi: 10.1242/jcs.199786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Cremer M, Kupper K, Wagler B, Wizelman L, von Hase J, Weiland Y, Kreja L, Diebold J, Speicher MR, Cremer T. Inheritance of gene density-related higher order chromatin arrangements in normal and tumor cell nuclei. The Journal of cell biology. 2003;162(5):809–20. doi: 10.1083/jcb.200304096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Leshner M, Devine M, Roloff GW, True LD, Misteli T, Meaburn KJ. Locus-specific gene repositioning in prostate cancer. Molecular biology of the cell. 2016;27(2):236–46. doi: 10.1091/mbc.E15-05-0280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Meaburn KJ, Gudla PR, Khan S, Lockett SJ, Misteli T. Disease-specific gene repositioning in breast cancer. The Journal of cell biology. 2009;187(6):801–12. doi: 10.1083/jcb.200909127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Meaburn KJ, Misteli T. Locus-specific and activity-independent gene repositioning during early tumorigenesis. J Cell Biol. 2008;180(1):39–50. doi: 10.1083/jcb.200708204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.De S, Michor F. DNA replication timing and long-range DNA interactions predict mutational landscapes of cancer genomes. Nature biotechnology. 2011;29(12):1103–8. doi: 10.1038/nbt.2030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Fudenberg G, Getz G, Meyerson M, Mirny LA. High order chromatin architecture shapes the landscape of chromosomal alterations in cancer. Nature biotechnology. 2011;29(12):1109–13. doi: 10.1038/nbt.2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Schuster-Bockler B, Lehner B. Chromatin organization is a major influence on regional mutation rates in human cancer cells. Nature. 2012;488(7412):504–7. doi: 10.1038/nature11273. [DOI] [PubMed] [Google Scholar]
- 155.Smith KS, Liu LL, Ganesan S, Michor F, De S. Nuclear topology modulates the mutational landscapes of cancer genomes. Nature structural & molecular biology. 2017;24(11):1000–1006. doi: 10.1038/nsmb.3474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Makova KD, Hardison RC. The effects of chromatin organization on variation in mutation rates in the genome. Nature reviews. Genetics. 2015;16(4):213–23. doi: 10.1038/nrg3890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Spielmann M, Mundlos S. Looking beyond the genes: the role of non-coding variants in human disease. Human molecular genetics. 2016;25(R2):R157–R165. doi: 10.1093/hmg/ddw205. [DOI] [PubMed] [Google Scholar]
- 158.Meddens CA, Harakalova M, van den Dungen NA, Foroughi Asl H, Hijma HJ, Cuppen EP, Bjorkegren JL, Asselbergs FW, Nieuwenhuis EE, Mokry M. Systematic analysis of chromatin interactions at disease associated loci links novel candidate genes to inflammatory bowel disease. Genome biology. 2016;17(1):247. doi: 10.1186/s13059-016-1100-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Brandt MM, Meddens CA, Louzao-Martinez L, van den Dungen NAM, Lansu NR, Nieuwenhuis EES, Duncker DJ, Verhaar MC, Joles JA, Mokry M, Cheng C. Chromatin Conformation Links Distal Target Genes to CKD Loci. Journal of the American Society of Nephrology : JASN. 2017 doi: 10.1681/ASN.2016080875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Haitjema S, Meddens CA, van der Laan SW, Kofink D, Harakalova M, Tragante V, Foroughi Asl H, van Setten J, Brandt MM, Bis JC, O'Donnell C, Cheng C, Hoefer IE, Waltenberger J, Biessen E, Jukema JW, Doevendans PA, Nieuwenhuis EE, Erdmann J, Bjorkegren JL, Pasterkamp G, Asselbergs FW, den Ruijter HM, Mokry M. Additional Candidate Genes for Human Atherosclerotic Disease Identified Through Annotation Based on Chromatin Organization. Circulation. Cardiovascular genetics. 2017;10(2) doi: 10.1161/CIRCGENETICS.116.001664. [DOI] [PubMed] [Google Scholar]
- 161.Burren OS, Rubio Garcia A, Javierre BM, Rainbow DB, Cairns J, Cooper NJ, Lambourne JJ, Schofield E, Castro Dopico X, Ferreira RC, Coulson R, Burden F, Rowlston SP, Downes K, Wingett SW, Frontini M, Ouwehand WH, Fraser P, Spivakov M, Todd JA, Wicker LS, Cutler AJ, Wallace C. Chromosome contacts in activated T cells identify autoimmune disease candidate genes. Genome biology. 2017;18(1):165. doi: 10.1186/s13059-017-1285-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Du M, Tillmans L, Gao J, Gao P, Yuan T, Dittmar RL, Song W, Yang Y, Sahr N, Wang T, Wei GH, Thibodeau SN, Wang L. Chromatin interactions and candidate genes at ten prostate cancer risk loci. Scientific reports. 2016;6:23202. doi: 10.1038/srep23202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Du M, Yuan T, Schilter KF, Dittmar RL, Mackinnon A, Huang X, Tschannen M, Worthey E, Jacob H, Xia S, Gao J, Tillmans L, Lu Y, Liu P, Thibodeau SN, Wang L. Prostate cancer risk locus at 8q24 as a regulatory hub by physical interactions with multiple genomic loci across the genome. Human molecular genetics. 2015;24(1):154–66. doi: 10.1093/hmg/ddu426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Kim K, Jang K, Yang W, Choi EY, Park SM, Bae M, Kim YJ, Choi JK. Chromatin structure-based prediction of recurrent noncoding mutations in cancer. Nature genetics. 2016;48(11):1321–1326. doi: 10.1038/ng.3682. [DOI] [PubMed] [Google Scholar]
- 165.Lettice LA, Daniels S, Sweeney E, Venkataraman S, Devenney PS, Gautier P, Morrison H, Fantes J, Hill RE, FitzPatrick DR. Enhancer-adoption as a mechanism of human developmental disease. Human mutation. 2011;32(12):1492–9. doi: 10.1002/humu.21615. [DOI] [PubMed] [Google Scholar]
- 166.Ibn-Salem J, Kohler S, Love MI, Chung HR, Huang N, Hurles ME, Haendel M, Washington NL, Smedley D, Mungall CJ, Lewis SE, Ott CE, Bauer S, Schofield PN, Mundlos S, Spielmann M, Robinson PN. Deletions of chromosomal regulatory boundaries are associated with congenital disease. Genome biology. 2014;15(9):423. doi: 10.1186/s13059-014-0423-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Northcott PA, Lee C, Zichner T, Stutz AM, Erkek S, Kawauchi D, Shih DJ, Hovestadt V, Zapatka M, Sturm D, Jones DT, Kool M, Remke M, Cavalli FM, Zuyderduyn S, Bader GD, VandenBerg S, Esparza LA, Ryzhova M, Wang W, Wittmann A, Stark S, Sieber L, Seker-Cin H, Linke L, Kratochwil F, Jager N, Buchhalter I, Imbusch CD, Zipprich G, Raeder B, Schmidt S, Diessl N, Wolf S, Wiemann S, Brors B, Lawerenz C, Eils J, Warnatz HJ, Risch T, Yaspo ML, Weber UD, Bartholomae CC, von Kalle C, Turanyi E, Hauser P, Sanden E, Darabi A, Siesjo P, Sterba J, Zitterbart K, Sumerauer D, van Sluis P, Versteeg R, Volckmann R, Koster J, Schuhmann MU, Ebinger M, Grimes HL, Robinson GW, Gajjar A, Mynarek M, von Hoff K, Rutkowski S, Pietsch T, Scheurlen W, Felsberg J, Reifenberger G, Kulozik AE, von Deimling A, Witt O, Eils R, Gilbertson RJ, Korshunov A, Taylor MD, Lichter P, Korbel JO, Wechsler-Reya RJ, Pfister SM. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature. 2014;511(7510):428–34. doi: 10.1038/nature13379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Yu M, Ren B. The Three-Dimensional Organization of Mammalian Genomes. Annual review of cell and developmental biology. 2017;33:265–289. doi: 10.1146/annurev-cellbio-100616-060531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Valton AL, Dekker J. TAD disruption as oncogenic driver. Current opinion in genetics & development. 2016;36:34–40. doi: 10.1016/j.gde.2016.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Lupianez DG, Spielmann M, Mundlos S. Breaking TADs: How Alterations of Chromatin Domains Result in Disease. Trends in genetics : TIG. 2016;32(4):225–37. doi: 10.1016/j.tig.2016.01.003. [DOI] [PubMed] [Google Scholar]
- 171.Kaiser VB, Semple CA. When TADs go bad: chromatin structure and nuclear organisation in human disease. F1000Research. 2017;6 doi: 10.12688/f1000research.10792.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Lupianez DG, Kraft K, Heinrich V, Krawitz P, Brancati F, Klopocki E, Horn D, Kayserili H, Opitz JM, Laxova R, Santos-Simarro F, Gilbert-Dussardier B, Wittler L, Borschiwer M, Haas SA, Osterwalder M, Franke M, Timmermann B, Hecht J, Spielmann M, Visel A, Mundlos S. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell. 2015;161(5):1012–1025. doi: 10.1016/j.cell.2015.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Katainen R, Dave K, Pitkanen E, Palin K, Kivioja T, Valimaki N, Gylfe AE, Ristolainen H, Hanninen UA, Cajuso T, Kondelin J, Tanskanen T, Mecklin JP, Jarvinen H, Renkonen-Sinisalo L, Lepisto A, Kaasinen E, Kilpivaara O, Tuupanen S, Enge M, Taipale J, Aaltonen LA. CTCF/cohesin-binding sites are frequently mutated in cancer. Nature genetics. 2015;47(7):818–21. doi: 10.1038/ng.3335. [DOI] [PubMed] [Google Scholar]
- 174.Flavahan WA, Drier Y, Liau BB, Gillespie SM, Venteicher AS, Stemmer-Rachamimov AO, Suva ML, Bernstein BE. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature. 2016;529(7584):110–4. doi: 10.1038/nature16490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Giorgio E, Robyr D, Spielmann M, Ferrero E, Di Gregorio E, Imperiale D, Vaula G, Stamoulis G, Santoni F, Atzori C, Gasparini L, Ferrera D, Canale C, Guipponi M, Pennacchio LA, Antonarakis SE, Brussino A, Brusco A. A large genomic deletion leads to enhancer adoption by the lamin B1 gene: a second path to autosomal dominant adult-onset demyelinating leukodystrophy (ADLD) Human molecular genetics. 2015;24(11):3143–54. doi: 10.1093/hmg/ddv065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Franke M, Ibrahim DM, Andrey G, Schwarzer W, Heinrich V, Schopflin R, Kraft K, Kempfer R, Jerkovic I, Chan WL, Spielmann M, Timmermann B, Wittler L, Kurth I, Cambiaso P, Zuffardi O, Houge G, Lambie L, Brancati F, Pombo A, Vingron M, Spitz F, Mundlos S. Formation of new chromatin domains determines pathogenicity of genomic duplications. Nature. 2016;538(7624):265–269. doi: 10.1038/nature19800. [DOI] [PubMed] [Google Scholar]
- 177.Groschel S, Sanders MA, Hoogenboezem R, de Wit E, Bouwman BAM, Erpelinck C, van der Velden VHJ, Havermans M, Avellino R, van Lom K, Rombouts EJ, van Duin M, Dohner K, Beverloo HB, Bradner JE, Dohner H, Lowenberg B, Valk PJM, Bindels EMJ, de Laat W, Delwel R. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell. 2014;157(2):369–381. doi: 10.1016/j.cell.2014.02.019. [DOI] [PubMed] [Google Scholar]
- 178.Weischenfeldt J, Dubash T, Drainas AP, Mardin BR, Chen Y, Stutz AM, Waszak SM, Bosco G, Halvorsen AR, Raeder B, Efthymiopoulos T, Erkek S, Siegl C, Brenner H, Brustugun OT, Dieter SM, Northcott PA, Petersen I, Pfister SM, Schneider M, Solberg SK, Thunissen E, Weichert W, Zichner T, Thomas R, Peifer M, Helland A, Ball CR, Jechlinger M, Sotillo R, Glimm H, Korbel JO. Pan-cancer analysis of somatic copy-number alterations implicates IRS4 and IGF2 in enhancer hijacking. Nature genetics. 2017;49(1):65–74. doi: 10.1038/ng.3722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Redin C, Brand H, Collins RL, Kammin T, Mitchell E, Hodge JC, Hanscom C, Pillalamarri V, Seabra CM, Abbott MA, Abdul-Rahman OA, Aberg E, Adley R, Alcaraz-Estrada SL, Alkuraya FS, An Y, Anderson MA, Antolik C, Anyane-Yeboa K, Atkin JF, Bartell T, Bernstein JA, Beyer E, Blumenthal I, Bongers EM, Brilstra EH, Brown CW, Bruggenwirth HT, Callewaert B, Chiang C, Corning K, Cox H, Cuppen E, Currall BB, Cushing T, David D, Deardorff MA, Dheedene A, D'Hooghe M, de Vries BB, Earl DL, Ferguson HL, Fisher H, FitzPatrick DR, Gerrol P, Giachino D, Glessner JT, Gliem T, Grady M, Graham BH, Griffis C, Gripp KW, Gropman AL, Hanson-Kahn A, Harris DJ, Hayden MA, Hill R, Hochstenbach R, Hoffman JD, Hopkin RJ, Hubshman MW, Innes AM, Irons M, Irving M, Jacobsen JC, Janssens S, Jewett T, Johnson JP, Jongmans MC, Kahler SG, Koolen DA, Korzelius J, Kroisel PM, Lacassie Y, Lawless W, Lemyre E, Leppig K, Levin AV, Li H, Li H, Liao EC, Lim C, Lose EJ, Lucente D, Macera MJ, Manavalan P, Mandrile G, Marcelis CL, Margolin L, Mason T, Masser-Frye D, McClellan MW, Mendoza CJ, Menten B, Middelkamp S, Mikami LR, Moe E, Mohammed S, Mononen T, Mortenson ME, Moya G, Nieuwint AW, Ordulu Z, Parkash S, Pauker SP, Pereira S, Perrin D, Phelan K, Aguilar RE, Poddighe PJ, Pregno G, Raskin S, Reis L, Rhead W, Rita D, Renkens I, Roelens F, Ruliera J, Rump P, Schilit SL, Shaheen R, Sparkes R, Spiegel E, Stevens B, Stone MR, Tagoe J, Thakuria JV, van Bon BW, van de Kamp J, van Der Burgt I, van Essen T, van Ravenswaaij-Arts CM, van Roosmalen MJ, Vergult S, Volker-Touw CM, Warburton DP, Waterman MJ, Wiley S, Wilson A, Yerena-de Vega MC, Zori RT, Levy B, Brunner HG, de Leeuw N, Kloosterman WP, Thorland EC, Morton CC, Gusella JF, Talkowski ME. The genomic landscape of balanced cytogenetic abnormalities associated with human congenital anomalies. Nature genetics. 2017;49(1):36–45. doi: 10.1038/ng.3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kaiser VB, Taylor MS, Semple CA. Mutational Biases Drive Elevated Rates of Substitution at Regulatory Sites across Cancer Types. PLoS genetics. 2016;12(8):e1006207. doi: 10.1371/journal.pgen.1006207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Hnisz D, Weintraub AS, Day DS, Valton AL, Bak RO, Li CH, Goldmann J, Lajoie BR, Fan ZP, Sigova AA, Reddy J, Borges-Rivera D, Lee TI, Jaenisch R, Porteus MH, Dekker J, Young RA. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science. 2016;351(6280):1454–1458. doi: 10.1126/science.aad9024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Dawson MA. The cancer epigenome: Concepts, challenges, and therapeutic opportunities. Science. 2017;355(6330):1147–1152. doi: 10.1126/science.aam7304. [DOI] [PubMed] [Google Scholar]
- 183.Taberlay PC, Achinger-Kawecka J, Lun AT, Buske FA, Sabir K, Gould CM, Zotenko E, Bert SA, Giles KA, Bauer DC, Smyth GK, Stirzaker C, O'Donoghue SI, Clark SJ. Three-dimensional disorganization of the cancer genome occurs coincident with long-range genetic and epigenetic alterations. Genome research. 2016;26(6):719–31. doi: 10.1101/gr.201517.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Wu P, Li T, Li R, Jia L, Zhu P, Liu Y, Chen Q, Tang D, Yu Y, Li C. 3D genome of multiple myeloma reveals spatial genome disorganization associated with copy number variations. Nature communications. 2017;8(1):1937. doi: 10.1038/s41467-017-01793-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Peifer M, Hertwig F, Roels F, Dreidax D, Gartlgruber M, Menon R, Kramer A, Roncaioli JL, Sand F, Heuckmann JM, Ikram F, Schmidt R, Ackermann S, Engesser A, Kahlert Y, Vogel W, Altmuller J, Nurnberg P, Thierry-Mieg J, Thierry-Mieg D, Mariappan A, Heynck S, Mariotti E, Henrich KO, Gloeckner C, Bosco G, Leuschner I, Schweiger MR, Savelyeva L, Watkins SC, Shao C, Bell E, Hofer T, Achter V, Lang U, Theissen J, Volland R, Saadati M, Eggert A, de Wilde B, Berthold F, Peng Z, Zhao C, Shi L, Ortmann M, Buttner R, Perner S, Hero B, Schramm A, Schulte JH, Herrmann C, O'Sullivan RJ, Westermann F, Thomas RK, Fischer M. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature. 2015;526(7575):700–4. doi: 10.1038/nature14980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Valentijn LJ, Koster J, Zwijnenburg DA, Hasselt NE, van Sluis P, Volckmann R, van Noesel MM, George RE, Tytgat GA, Molenaar JJ, Versteeg R. TERT rearrangements are frequent in neuroblastoma and identify aggressive tumors. Nature genetics. 2015;47(12):1411–4. doi: 10.1038/ng.3438. [DOI] [PubMed] [Google Scholar]
- 187.Borden J, Manuelidis L. Movement of the X chromosome in epilepsy. Science. 1988;242(4886):1687–91. doi: 10.1126/science.3201257. [DOI] [PubMed] [Google Scholar]
- 188.Walczak A, Szczepankiewicz AA, Ruszczycki B, Magalska A, Zamlynska K, Dzwonek J, Wilczek E, Zybura-Broda K, Rylski M, Malinowska M, Dabrowski M, Szczepinska T, Pawlowski K, Pyskaty M, Wlodarczyk J, Szczerbal I, Switonski M, Cremer M, Wilczynski GM. Novel higher-order epigenetic regulation of the Bdnf gene upon seizures. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2013;33(6):2507–11. doi: 10.1523/JNEUROSCI.1085-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Gregor A, Oti M, Kouwenhoven EN, Hoyer J, Sticht H, Ekici AB, Kjaergaard S, Rauch A, Stunnenberg HG, Uebe S, Vasileiou G, Reis A, Zhou H, Zweier C. De novo mutations in the genome organizer CTCF cause intellectual disability. American journal of human genetics. 2013;93(1):124–31. doi: 10.1016/j.ajhg.2013.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Bastaki F, Nair P, Mohamed M, Malik EM, Helmi M, Al-Ali MT, Hamzeh AR. Identification of a novel CTCF mutation responsible for syndromic intellectual disability - a case report. BMC medical genetics. 2017;18(1):68. doi: 10.1186/s12881-017-0429-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Kennedy JA, Ebert BL. Clinical Implications of Genetic Mutations in Myelodysplastic Syndrome. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(9):968–974. doi: 10.1200/JCO.2016.71.0806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Liu J, Krantz ID. Cohesin and human disease. Annual review of genomics and human genetics. 2008;9:303–20. doi: 10.1146/annurev.genom.9.081307.164211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Zakari M, Yuen K, Gerton JL. Etiology and pathogenesis of the cohesinopathies. Wiley interdisciplinary reviews. Developmental biology. 2015;4(5):489–504. doi: 10.1002/wdev.190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Banerji R, Skibbens RV, Iovine MK. How many roads lead to cohesinopathies? Developmental dynamics : an official publication of the American Association of Anatomists. 2017;246(11):881–888. doi: 10.1002/dvdy.24510. [DOI] [PubMed] [Google Scholar]
- 195.Capell BC, Collins FS. Human laminopathies: nuclei gone genetically awry. Nature reviews. Genetics. 2006;7(12):940–52. doi: 10.1038/nrg1906. [DOI] [PubMed] [Google Scholar]
- 196.Worman HJ, Bonne G. "Laminopathies": a wide spectrum of human diseases. Experimental cell research. 2007;313(10):2121–33. doi: 10.1016/j.yexcr.2007.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Dekker J, Belmont AS, Guttman M, Leshyk VO, Lis JT, Lomvardas S, Mirny LA, O'Shea CC, Park PJ, Ren B, Politz JCR, Shendure J, Zhong S, Network DN. The 4D nucleome project. Nature. 2017;549(7671):219–226. doi: 10.1038/nature23884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Nagano T, Lubling Y, Stevens TJ, Schoenfelder S, Yaffe E, Dean W, Laue ED, Tanay A, Fraser P. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature. 2013;502(7469):59–64. doi: 10.1038/nature12593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Ramani V, Deng X, Qiu R, Gunderson KL, Steemers FJ, Disteche CM, Noble WS, Duan Z, Shendure J. Massively multiplex single-cell Hi-C. Nature methods. 2017;14(3):263–266. doi: 10.1038/nmeth.4155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Carter D, Chakalova L, Osborne CS, Dai YF, Fraser P. Long-range chromatin regulatory interactions in vivo. Nature genetics. 2002;32(4):623–6. doi: 10.1038/ng1051. [DOI] [PubMed] [Google Scholar]
- 201.Beagrie RA, Scialdone A, Schueler M, Kraemer DC, Chotalia M, Xie SQ, Barbieri M, de Santiago I, Lavitas LM, Branco MR, Fraser J, Dostie J, Game L, Dillon N, Edwards PA, Nicodemi M, Pombo A. Complex multi-enhancer contacts captured by genome architecture mapping. Nature. 2017;543(7646):519–524. doi: 10.1038/nature21411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Quinodoz S, Ollikainen N, Tabak B, Palla A, Schmidt J, Detmar E, Lai M, Shishkin A, Bhat P, Trinh V, Aznauryan E, Russell P, Cheng C, Jovanovic M, Chow A, McDonel P, Garber M, Guttman M. Higher-order inter-chromosomal hubs shape 3-dimensional genome organization in the nucleus. BioRxiv. 2017 doi: 10.1016/j.cell.2018.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Zheng M, Tian SZ, Maurya R, Lee B, Kim M, Capurso D, Piecuch E, Gong L, Zhu JJ, Wong CH, Ngan CY, Wang P, Ruan X, Wei C-L, Ruan Y. Multiplex Chromatin Interaction Analysis with Single-Molecule Precision. bioRxiv. 2018 doi: 10.1038/s41586-019-0949-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Liu X, Zhang Y, Chen Y, Li M, Zhou F, Li K, Cao H, Ni M, Liu Y, Gu Z, Dickerson KE, Xie S, Hon GC, Xuan Z, Zhang MQ, Shao Z, Xu J. In Situ Capture of Chromatin Interactions by Biotinylated dCas9. Cell. 2017;170(5):1028–1043. e19. doi: 10.1016/j.cell.2017.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Dekker J, Rippe K, Dekker M, Kleckner N. Capturing chromosome conformation. Science. 2002;295(5558):1306–11. doi: 10.1126/science.1067799. [DOI] [PubMed] [Google Scholar]
- 206.Zhao Z, Tavoosidana G, Sjolinder M, Gondor A, Mariano P, Wang S, Kanduri C, Lezcano M, Sandhu KS, Singh U, Pant V, Tiwari V, Kurukuti S, Ohlsson R. Circular chromosome conformation capture (4C) uncovers extensive networks of epigenetically regulated intra- and interchromosomal interactions. Nature genetics. 2006;38(11):1341–7. doi: 10.1038/ng1891. [DOI] [PubMed] [Google Scholar]
- 207.Simonis M, Klous P, Splinter E, Moshkin Y, Willemsen R, de Wit E, van Steensel B, de Laat W. Nuclear organization of active and inactive chromatin domains uncovered by chromosome conformation capture-on-chip (4C) Nature genetics. 2006;38(11):1348–54. doi: 10.1038/ng1896. [DOI] [PubMed] [Google Scholar]
- 208.Schoenfelder S, Sexton T, Chakalova L, Cope NF, Horton A, Andrews S, Kurukuti S, Mitchell JA, Umlauf D, Dimitrova DS, Eskiw CH, Luo Y, Wei CL, Ruan Y, Bieker JJ, Fraser P. Preferential associations between co-regulated genes reveal a transcriptional interactome in erythroid cells. Nature genetics. 2010;42(1):53–61. doi: 10.1038/ng.496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Schwartzman O, Mukamel Z, Oded-Elkayam N, Olivares-Chauvet P, Lubling Y, Landan G, Izraeli S, Tanay A. UMI-4C for quantitative and targeted chromosomal contact profiling. Nature methods. 2016;13(8):685–91. doi: 10.1038/nmeth.3922. [DOI] [PubMed] [Google Scholar]
- 210.Ling JQ, Li T, Hu JF, Vu TH, Chen HL, Qiu XW, Cherry AM, Hoffman AR. CTCF mediates interchromosomal colocalization between Igf2/H19 and Wsb1/Nf1. Science. 2006;312(5771):269–72. doi: 10.1126/science.1123191. [DOI] [PubMed] [Google Scholar]
- 211.Dostie J, Richmond TA, Arnaout RA, Selzer RR, Lee WL, Honan TA, Rubio ED, Krumm A, Lamb J, Nusbaum C, Green RD, Dekker J. Chromosome Conformation Capture Carbon Copy (5C): a massively parallel solution for mapping interactions between genomic elements. Genome research. 2006;16(10):1299–309. doi: 10.1101/gr.5571506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Hughes JR, Roberts N, McGowan S, Hay D, Giannoulatou E, Lynch M, De Gobbi M, Taylor S, Gibbons R, Higgs DR. Analysis of hundreds of cis-regulatory landscapes at high resolution in a single, high-throughput experiment. Nature genetics. 2014;46(2):205–12. doi: 10.1038/ng.2871. [DOI] [PubMed] [Google Scholar]
- 213.Mifsud B, Tavares-Cadete F, Young AN, Sugar R, Schoenfelder S, Ferreira L, Wingett SW, Andrews S, Grey W, Ewels PA, Herman B, Happe S, Higgs A, LeProust E, Follows GA, Fraser P, Luscombe NM, Osborne CS. Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C. Nature genetics. 2015;47(6):598–606. doi: 10.1038/ng.3286. [DOI] [PubMed] [Google Scholar]
- 214.Fullwood MJ, Liu MH, Pan YF, Liu J, Xu H, Mohamed YB, Orlov YL, Velkov S, Ho A, Mei PH, Chew EG, Huang PY, Welboren WJ, Han Y, Ooi HS, Ariyaratne PN, Vega VB, Luo Y, Tan PY, Choy PY, Wansa KD, Zhao B, Lim KS, Leow SC, Yow JS, Joseph R, Li H, Desai KV, Thomsen JS, Lee YK, Karuturi RK, Herve T, Bourque G, Stunnenberg HG, Ruan X, Cacheux-Rataboul V, Sung WK, Liu ET, Wei CL, Cheung E, Ruan Y. An oestrogen-receptor-alpha-bound human chromatin interactome. Nature. 2009;462(7269):58–64. doi: 10.1038/nature08497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Mumbach MR, Rubin AJ, Flynn RA, Dai C, Khavari PA, Greenleaf WJ, Chang HY. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nature methods. 2016;13(11):919–922. doi: 10.1038/nmeth.3999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Fang R, Yu M, Li G, Chee S, Liu T, Schmitt AD, Ren B. Mapping of long-range chromatin interactions by proximity ligation-assisted ChIP-seq. Cell research. 2016;26(12):1345–1348. doi: 10.1038/cr.2016.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Kalhor R, Tjong H, Jayathilaka N, Alber F, Chen L. Genome architectures revealed by tethered chromosome conformation capture and population-based modeling. Nature biotechnology. 2012;30(1):90–8. doi: 10.1038/nbt.2057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Hsieh TH, Weiner A, Lajoie B, Dekker J, Friedman N, Rando OJ. Mapping Nucleosome Resolution Chromosome Folding in Yeast by Micro-C. Cell. 2015;162(1):108–19. doi: 10.1016/j.cell.2015.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Olivares-Chauvet P, Mukamel Z, Lifshitz A, Schwartzman O, Elkayam NO, Lubling Y, Deikus G, Sebra RP, Tanay A. Capturing pairwise and multi-way chromosomal conformations using chromosomal walks. Nature. 2016;540(7632):296–300. doi: 10.1038/nature20158. [DOI] [PubMed] [Google Scholar]
- 220.Liang Z, Li G, Wang Z, Djekidel MN, Li Y, Qian MP, Zhang MQ, Chen Y. BL-Hi-C is an efficient and sensitive approach for capturing structural and regulatory chromatin interactions. Nature communications. 2017;8(1):1622. doi: 10.1038/s41467-017-01754-3. [DOI] [PMC free article] [PubMed] [Google Scholar]



