Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 415
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(18): 4926-4945.e22, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38986619

ABSTRACT

Posterior fossa group A (PFA) ependymoma is a lethal brain cancer diagnosed in infants and young children. The lack of driver events in the PFA linear genome led us to search its 3D genome for characteristic features. Here, we reconstructed 3D genomes from diverse childhood tumor types and uncovered a global topology in PFA that is highly reminiscent of stem and progenitor cells in a variety of human tissues. A remarkable feature exclusively present in PFA are type B ultra long-range interactions in PFAs (TULIPs), regions separated by great distances along the linear genome that interact with each other in the 3D nuclear space with surprising strength. TULIPs occur in all PFA samples and recur at predictable genomic coordinates, and their formation is induced by expression of EZHIP. The universality of TULIPs across PFA samples suggests a conservation of molecular principles that could be exploited therapeutically.


Subject(s)
Ependymoma , Ependymoma/genetics , Humans , Infratentorial Neoplasms/genetics , Infratentorial Neoplasms/pathology , Genome, Human , Infant , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Male , Female
2.
Cell ; 186(20): 4404-4421.e20, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37774679

ABSTRACT

Persistent DNA double-strand breaks (DSBs) in neurons are an early pathological hallmark of neurodegenerative diseases including Alzheimer's disease (AD), with the potential to disrupt genome integrity. We used single-nucleus RNA-seq in human postmortem prefrontal cortex samples and found that excitatory neurons in AD were enriched for somatic mosaic gene fusions. Gene fusions were particularly enriched in excitatory neurons with DNA damage repair and senescence gene signatures. In addition, somatic genome structural variations and gene fusions were enriched in neurons burdened with DSBs in the CK-p25 mouse model of neurodegeneration. Neurons enriched for DSBs also had elevated levels of cohesin along with progressive multiscale disruption of the 3D genome organization aligned with transcriptional changes in synaptic, neuronal development, and histone genes. Overall, this study demonstrates the disruption of genome stability and the 3D genome organization by DSBs in neurons as pathological steps in the progression of neurodegenerative diseases.


Subject(s)
DNA Breaks, Double-Stranded , Neurodegenerative Diseases , Animals , Humans , Mice , Alzheimer Disease/genetics , DNA , DNA Repair/genetics , Neurodegenerative Diseases/genetics , Neurons/physiology , Single-Cell Analysis , Sequence Analysis, RNA , Genomic Instability
3.
Cell ; 186(24): 5269-5289.e22, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37995656

ABSTRACT

A generic level of chromatin organization generated by the interplay between cohesin and CTCF suffices to limit promiscuous interactions between regulatory elements, but a lineage-specific chromatin assembly that supersedes these constraints is required to configure the genome to guide gene expression changes that drive faithful lineage progression. Loss-of-function approaches in B cell precursors show that IKAROS assembles interactions across megabase distances in preparation for lymphoid development. Interactions emanating from IKAROS-bound enhancers override CTCF-imposed boundaries to assemble lineage-specific regulatory units built on a backbone of smaller invariant topological domains. Gain of function in epithelial cells confirms IKAROS' ability to reconfigure chromatin architecture at multiple scales. Although the compaction of the Igκ locus required for genome editing represents a function of IKAROS unique to lymphocytes, the more general function to preconfigure the genome to support lineage-specific gene expression and suppress activation of extra-lineage genes provides a paradigm for lineage restriction.


Subject(s)
Chromatin , Genome , B-Lymphocytes/metabolism , CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , Humans , Animals , Mice
4.
Cell ; 185(20): 3689-3704.e21, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179666

ABSTRACT

Regulatory landscapes drive complex developmental gene expression, but it remains unclear how their integrity is maintained when incorporating novel genes and functions during evolution. Here, we investigated how a placental mammal-specific gene, Zfp42, emerged in an ancient vertebrate topologically associated domain (TAD) without adopting or disrupting the conserved expression of its gene, Fat1. In ESCs, physical TAD partitioning separates Zfp42 and Fat1 with distinct local enhancers that drive their independent expression. This separation is driven by chromatin activity and not CTCF/cohesin. In contrast, in embryonic limbs, inactive Zfp42 shares Fat1's intact TAD without responding to active Fat1 enhancers. However, neither Fat1 enhancer-incompatibility nor nuclear envelope-attachment account for Zfp42's unresponsiveness. Rather, Zfp42's promoter is rendered inert to enhancers by context-dependent DNA methylation. Thus, diverse mechanisms enabled the integration of independent Zfp42 regulation in the Fat1 locus. Critically, such regulatory complexity appears common in evolution as, genome wide, most TADs contain multiple independently expressed genes.


Subject(s)
Chromatin , Placenta , Animals , CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Evolution, Molecular , Female , Genome , Mammals/metabolism , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cell ; 184(3): 723-740.e21, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33508230

ABSTRACT

Elucidating the regulatory mechanisms of human brain evolution is essential to understanding human cognition and mental disorders. We generated multi-omics profiles and constructed a high-resolution map of 3D genome architecture of rhesus macaque during corticogenesis. By comparing the 3D genomes of human, macaque, and mouse brains, we identified many human-specific chromatin structure changes, including 499 topologically associating domains (TADs) and 1,266 chromatin loops. The human-specific loops are significantly enriched in enhancer-enhancer interactions, and the regulated genes show human-specific expression changes in the subplate, a transient zone of the developing brain critical for neural circuit formation and plasticity. Notably, many human-specific sequence changes are located in the human-specific TAD boundaries and loop anchors, which may generate new transcription factor binding sites and chromatin structures in human. Collectively, the presented data highlight the value of comparative 3D genome analyses in dissecting the regulatory mechanisms of brain development and evolution.


Subject(s)
Brain/embryology , Evolution, Molecular , Fetus/embryology , Genome , Organogenesis/genetics , Animals , Base Sequence , Chromatin/metabolism , DNA Transposable Elements/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Humans , Macaca mulatta , Mice , Species Specificity , Synteny/genetics , Transcription Factors/metabolism
6.
Cell ; 184(25): 6157-6173.e24, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34856126

ABSTRACT

Chromosome loops shift dynamically during development, homeostasis, and disease. CCCTC-binding factor (CTCF) is known to anchor loops and construct 3D genomes, but how anchor sites are selected is not yet understood. Here, we unveil Jpx RNA as a determinant of anchor selectivity. Jpx RNA targets thousands of genomic sites, preferentially binding promoters of active genes. Depleting Jpx RNA causes ectopic CTCF binding, massive shifts in chromosome looping, and downregulation of >700 Jpx target genes. Without Jpx, thousands of lost loops are replaced by de novo loops anchored by ectopic CTCF sites. Although Jpx controls CTCF binding on a genome-wide basis, it acts selectively at the subset of developmentally sensitive CTCF sites. Specifically, Jpx targets low-affinity CTCF motifs and displaces CTCF protein through competitive inhibition. We conclude that Jpx acts as a CTCF release factor and shapes the 3D genome by regulating anchor site usage.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromosomes/metabolism , RNA, Long Noncoding/metabolism , Animals , Binding Sites , Cell Line , Embryonic Stem Cells , Mice , Protein Binding
7.
Cell ; 182(6): 1641-1659.e26, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32822575

ABSTRACT

The 3D organization of chromatin regulates many genome functions. Our understanding of 3D genome organization requires tools to directly visualize chromatin conformation in its native context. Here we report an imaging technology for visualizing chromatin organization across multiple scales in single cells with high genomic throughput. First we demonstrate multiplexed imaging of hundreds of genomic loci by sequential hybridization, which allows high-resolution conformation tracing of whole chromosomes. Next we report a multiplexed error-robust fluorescence in situ hybridization (MERFISH)-based method for genome-scale chromatin tracing and demonstrate simultaneous imaging of more than 1,000 genomic loci and nascent transcripts of more than 1,000 genes together with landmark nuclear structures. Using this technology, we characterize chromatin domains, compartments, and trans-chromosomal interactions and their relationship to transcription in single cells. We envision broad application of this high-throughput, multi-scale, and multi-modal imaging technology, which provides an integrated view of chromatin organization in its native structural and functional context.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Chromosomes, Human/metabolism , High-Throughput Screening Assays/methods , In Situ Hybridization, Fluorescence/methods , Single-Cell Analysis/methods , Algorithms , Cell Line , Cell Nucleus/genetics , Chromatin/genetics , Chromosomes, Human/genetics , DNA/genetics , DNA/metabolism , Genomics , Humans , Image Processing, Computer-Assisted , Molecular Conformation , Multimodal Imaging , Nucleolus Organizer Region/genetics , Nucleolus Organizer Region/metabolism , RNA/genetics , RNA/metabolism , Software
8.
Cell ; 175(5): 1405-1417.e14, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30318144

ABSTRACT

Programmable control of spatial genome organization is a powerful approach for studying how nuclear structure affects gene regulation and cellular function. Here, we develop a versatile CRISPR-genome organization (CRISPR-GO) system that can efficiently control the spatial positioning of genomic loci relative to specific nuclear compartments, including the nuclear periphery, Cajal bodies, and promyelocytic leukemia (PML) bodies. CRISPR-GO is chemically inducible and reversible, enabling interrogation of real-time dynamics of chromatin interactions with nuclear compartments in living cells. Inducible repositioning of genomic loci to the nuclear periphery allows for dissection of mitosis-dependent and -independent relocalization events and also for interrogation of the relationship between gene position and gene expression. CRISPR-GO mediates rapid de novo formation of Cajal bodies at desired chromatin loci and causes significant repression of endogenous gene expression over long distances (30-600 kb). The CRISPR-GO system offers a programmable platform to investigate large-scale spatial genome organization and function.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Genome , Abscisic Acid/pharmacology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Coiled Bodies/genetics , Gene Expression Regulation , Genetic Loci , Humans , In Situ Hybridization, Fluorescence , S Phase Cell Cycle Checkpoints/drug effects
9.
Cell ; 175(1): 224-238.e15, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30173918

ABSTRACT

More than 25 inherited human disorders are caused by the unstable expansion of repetitive DNA sequences termed short tandem repeats (STRs). A fundamental unresolved question is why some STRs are susceptible to pathologic expansion, whereas thousands of repeat tracts across the human genome are relatively stable. Here, we discover that nearly all disease-associated STRs (daSTRs) are located at boundaries demarcating 3D chromatin domains. We identify a subset of boundaries with markedly higher CpG island density compared to the rest of the genome. daSTRs specifically localize to ultra-high-density CpG island boundaries, suggesting they might be hotspots for epigenetic misregulation or topological disruption linked to STR expansion. Fragile X syndrome patients exhibit severe boundary disruption in a manner that correlates with local loss of CTCF occupancy and the degree of FMR1 silencing. Our data uncover higher-order chromatin architecture as a new dimension in understanding repeat expansion disorders.


Subject(s)
Chromatin/genetics , Microsatellite Repeats/physiology , Trinucleotide Repeat Expansion/physiology , Adult , Brain/cytology , Brain/pathology , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/physiology , Cell Line , Chromatin/physiology , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , CpG Islands/genetics , CpG Islands/physiology , DNA/genetics , Disease/etiology , Disease/genetics , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/physiology , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Genome, Human/genetics , Humans , Male , Microsatellite Repeats/genetics , Trinucleotide Repeat Expansion/genetics
10.
Annu Rev Cell Dev Biol ; 35: 357-379, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31283382

ABSTRACT

Eukaryotic transcription factors (TFs) from the same structural family tend to bind similar DNA sequences, despite the ability of these TFs to execute distinct functions in vivo. The cell partly resolves this specificity paradox through combinatorial strategies and the use of low-affinity binding sites, which are better able to distinguish between similar TFs. However, because these sites have low affinity, it is challenging to understand how TFs recognize them in vivo. Here, we summarize recent findings and technological advancements that allow for the quantification and mechanistic interpretation of TF recognition across a wide range of affinities. We propose a model that integrates insights from the fields of genetics and cell biology to provide further conceptual understanding of TF binding specificity. We argue that in eukaryotes, target specificity is driven by an inhomogeneous 3D nuclear distribution of TFs and by variation in DNA binding affinity such that locally elevated TF concentration allows low-affinity binding sites to be functional.


Subject(s)
Eukaryota/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism , Animals , Binding Sites , Gene Expression Regulation , Humans
11.
Immunity ; 56(7): 1451-1467.e12, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37263273

ABSTRACT

Multi-enhancer hubs are spatial clusters of enhancers present across numerous developmental programs. Here, we studied the functional relevance of these three-dimensional structures in T cell biology. Mathematical modeling identified a highly connected multi-enhancer hub at the Ets1 locus, comprising a noncoding regulatory element that was a hotspot for sequence variation associated with allergic disease in humans. Deletion of this regulatory element in mice revealed that the multi-enhancer connectivity was dispensable for T cell development but required for CD4+ T helper 1 (Th1) differentiation. These mice were protected from Th1-mediated colitis but exhibited overt allergic responses. Mechanistically, the multi-enhancer hub controlled the dosage of Ets1 that was required for CTCF recruitment and assembly of Th1-specific genome topology. Our findings establish a paradigm wherein multi-enhancer hubs control cellular competence to respond to an inductive cue through quantitative control of gene dosage and provide insight into how sequence variation within noncoding elements at the Ets1 locus predisposes individuals to allergic responses.


Subject(s)
Hypersensitivity , T-Lymphocytes , Humans , Mice , Animals , Cell Differentiation/genetics , Hematopoiesis , Inflammation/genetics , Regulatory Sequences, Nucleic Acid , Hypersensitivity/genetics , Enhancer Elements, Genetic/genetics
12.
Cell ; 171(3): 557-572.e24, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29053968

ABSTRACT

Chromosome conformation capture technologies have revealed important insights into genome folding. Yet, how spatial genome architecture is related to gene expression and cell fate remains unclear. We comprehensively mapped 3D chromatin organization during mouse neural differentiation in vitro and in vivo, generating the highest-resolution Hi-C maps available to date. We found that transcription is correlated with chromatin insulation and long-range interactions, but dCas9-mediated activation is insufficient for creating TAD boundaries de novo. Additionally, we discovered long-range contacts between gene bodies of exon-rich, active genes in all cell types. During neural differentiation, contacts between active TADs become less pronounced while inactive TADs interact more strongly. An extensive Polycomb network in stem cells is disrupted, while dynamic interactions between neural transcription factors appear in vivo. Finally, cell type-specific enhancer-promoter contacts are established concomitant to gene expression. This work shows that multiple factors influence the dynamics of chromatin interactions in development.


Subject(s)
Chromatin/metabolism , Genome , Neurogenesis , Animals , CCCTC-Binding Factor , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Exons , Gene Expression , Gene Regulatory Networks , Mice , Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription Factors/metabolism
13.
Mol Cell ; 84(11): 2017-2035.e6, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38795706

ABSTRACT

Whether and how histone post-translational modifications and the proteins that bind them drive 3D genome organization remains unanswered. Here, we evaluate the contribution of H3K9-methylated constitutive heterochromatin to 3D genome organization in Drosophila tissues. We find that the predominant organizational feature of wild-type tissues is the segregation of euchromatic chromosome arms from heterochromatic pericentromeres. Reciprocal perturbation of HP1a⋅H3K9me binding, using a point mutation in the HP1a chromodomain or replacement of the replication-dependent histone H3 with H3K9R mutant histones, revealed that HP1a binding to methylated H3K9 in constitutive heterochromatin is required to limit contact frequency between pericentromeres and chromosome arms and regulate the distance between arm and pericentromeric regions. Surprisingly, the self-association of pericentromeric regions is largely preserved despite the loss of H3K9 methylation and HP1a occupancy. Thus, the HP1a⋅H3K9 interaction contributes to but does not solely drive the segregation of euchromatin and heterochromatin inside the nucleus.


Subject(s)
Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone , Drosophila Proteins , Drosophila melanogaster , Heterochromatin , Histones , Heterochromatin/metabolism , Heterochromatin/genetics , Animals , Histones/metabolism , Histones/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Methylation , Euchromatin/metabolism , Euchromatin/genetics , Centromere/metabolism , Centromere/genetics , Protein Binding , Genome, Insect , Chromosome Segregation , Protein Processing, Post-Translational
14.
Mol Cell ; 84(4): 621-639.e9, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38244545

ABSTRACT

The DNA-binding protein SATB2 is genetically linked to human intelligence. We studied its influence on the three-dimensional (3D) epigenome by mapping chromatin interactions and accessibility in control versus SATB2-deficient cortical neurons. We find that SATB2 affects the chromatin looping between enhancers and promoters of neuronal-activity-regulated genes, thus influencing their expression. It also alters A/B compartments, topologically associating domains, and frequently interacting regions. Genes linked to SATB2-dependent 3D genome changes are implicated in highly specialized neuronal functions and contribute to cognitive ability and risk for neuropsychiatric and neurodevelopmental disorders. Non-coding DNA regions with a SATB2-dependent structure are enriched for common variants associated with educational attainment, intelligence, and schizophrenia. Our data establish SATB2 as a cell-type-specific 3D genome modulator, which operates both independently and in cooperation with CCCTC-binding factor (CTCF) to set up the chromatin landscape of pyramidal neurons for cognitive processes.


Subject(s)
Matrix Attachment Region Binding Proteins , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Neurons/metabolism , CCCTC-Binding Factor/metabolism , Chromatin/genetics , Chromatin/metabolism , Genome , Cognition , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism
15.
Mol Cell ; 84(8): 1406-1421.e8, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38490199

ABSTRACT

Enhancers bind transcription factors, chromatin regulators, and non-coding transcripts to modulate the expression of target genes. Here, we report 3D genome structures of single mouse ES cells as they are induced to exit pluripotency and transition through a formative stage prior to undergoing neuroectodermal differentiation. We find that there is a remarkable reorganization of 3D genome structure where inter-chromosomal intermingling increases dramatically in the formative state. This intermingling is associated with the formation of a large number of multiway hubs that bring together enhancers and promoters with similar chromatin states from typically 5-8 distant chromosomal sites that are often separated by many Mb from each other. In the formative state, genes important for pluripotency exit establish contacts with emerging enhancers within these multiway hubs, suggesting that the structural changes we have observed may play an important role in modulating transcription and establishing new cell identities.


Subject(s)
Mouse Embryonic Stem Cells , Regulatory Sequences, Nucleic Acid , Mice , Animals , Mouse Embryonic Stem Cells/metabolism , Embryonic Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic
16.
Mol Cell ; 83(19): 3457-3469.e7, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37802023

ABSTRACT

Circadian gene transcription is fundamental to metabolic physiology. Here we report that the nuclear receptor REV-ERBα, a repressive component of the molecular clock, forms circadian condensates in the nuclei of mouse liver. These condensates are dictated by an intrinsically disordered region (IDR) located in the protein's hinge region which specifically concentrates nuclear receptor corepressor 1 (NCOR1) at the genome. IDR deletion diminishes the recruitment of NCOR1 and disrupts rhythmic gene transcription in vivo. REV-ERBα condensates are located at high-order transcriptional repressive hubs in the liver genome that are highly correlated with circadian gene repression. Deletion of the IDR disrupts transcriptional repressive hubs and diminishes silencing of target genes by REV-ERBα. This work demonstrates physiological circadian protein condensates containing REV-ERBα whose IDR is required for hub formation and the control of rhythmic gene expression.


Subject(s)
Circadian Clocks , Mice , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Liver/metabolism , Gene Expression
17.
Mol Cell ; 83(9): 1446-1461.e6, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36996812

ABSTRACT

Enhancer clusters overlapping disease-associated mutations in Pierre Robin sequence (PRS) patients regulate SOX9 expression at genomic distances over 1.25 Mb. We applied optical reconstruction of chromatin architecture (ORCA) imaging to trace 3D locus topology during PRS-enhancer activation. We observed pronounced changes in locus topology between cell types. Subsequent analysis of single-chromatin fiber traces revealed that these ensemble-average differences arise through changes in the frequency of commonly sampled topologies. We further identified two CTCF-bound elements, internal to the SOX9 topologically associating domain, which promote stripe formation, are positioned near the domain's 3D geometric center, and bridge enhancer-promoter contacts in a series of chromatin loops. Ablation of these elements results in diminished SOX9 expression and altered domain-wide contacts. Polymer models with uniform loading across the domain and frequent cohesin collisions recapitulate this multi-loop, centrally clustered geometry. Together, we provide mechanistic insights into architectural stripe formation and gene regulation over ultra-long genomic ranges.


Subject(s)
Chromatin , Regulatory Sequences, Nucleic Acid , Humans , Chromatin/genetics , Promoter Regions, Genetic , Gene Expression Regulation , Genome , Cell Cycle Proteins/metabolism , Enhancer Elements, Genetic , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism
18.
Mol Cell ; 83(16): 2856-2871.e8, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37536339

ABSTRACT

Cohesin and CCCTC-binding factor (CTCF) are key regulatory proteins of three-dimensional (3D) genome organization. Cohesin extrudes DNA loops that are anchored by CTCF in a polar orientation. Here, we present direct evidence that CTCF binding polarity controls cohesin-mediated DNA looping. Using single-molecule imaging, we demonstrate that a critical N-terminal motif of CTCF blocks cohesin translocation and DNA looping. The cryo-EM structure of the cohesin-CTCF complex reveals that this CTCF motif ahead of zinc fingers can only reach its binding site on the STAG1 cohesin subunit when the N terminus of CTCF faces cohesin. Remarkably, a C-terminally oriented CTCF accelerates DNA compaction by cohesin. DNA-bound Cas9 and Cas12a ribonucleoproteins are also polar cohesin barriers, indicating that stalling may be intrinsic to cohesin itself. Finally, we show that RNA-DNA hybrids (R-loops) block cohesin-mediated DNA compaction in vitro and are enriched with cohesin subunits in vivo, likely forming TAD boundaries.


Subject(s)
Chromatin , R-Loop Structures , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA/genetics , DNA/metabolism , Cohesins
19.
Mol Cell ; 83(9): 1377-1392.e6, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37146570

ABSTRACT

Although population-level analyses revealed significant roles for CTCF and cohesin in mammalian genome organization, their contributions at the single-cell level remain incompletely understood. Here, we used a super-resolution microscopy approach to measure the effects of removal of CTCF or cohesin in mouse embryonic stem cells. Single-chromosome traces revealed cohesin-dependent loops, frequently stacked at their loop anchors forming multi-way contacts (hubs), bridging across TAD boundaries. Despite these bridging interactions, chromatin in intervening TADs was not intermixed, remaining separated in distinct loops around the hub. At the multi-TAD scale, steric effects from loop stacking insulated local chromatin from ultra-long range (>4 Mb) contacts. Upon cohesin removal, the chromosomes were more disordered and increased cell-cell variability in gene expression. Our data revise the TAD-centric understanding of CTCF and cohesin and provide a multi-scale, structural picture of how they organize the genome on the single-cell level through distinct contributions to loop stacking.


Subject(s)
Chromatin , Chromosomes , Animals , Mice , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Chromosomes/genetics , Chromosomes/metabolism , Chromatin/genetics , Chromatin/metabolism , Mouse Embryonic Stem Cells/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Mammals/metabolism
20.
Mol Cell ; 82(20): 3769-3780.e5, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36182691

ABSTRACT

Complex genomes show intricate organization in three-dimensional (3D) nuclear space. Current models posit that cohesin extrudes loops to form self-interacting domains delimited by the DNA binding protein CTCF. Here, we describe and quantitatively characterize cohesin-propelled, jet-like chromatin contacts as landmarks of loop extrusion in quiescent mammalian lymphocytes. Experimental observations and polymer simulations indicate that narrow origins of loop extrusion favor jet formation. Unless constrained by CTCF, jets propagate symmetrically for 1-2 Mb, providing an estimate for the range of in vivo loop extrusion. Asymmetric CTCF binding deflects the angle of jet propagation as experimental evidence that cohesin-mediated loop extrusion can switch from bi- to unidirectional and is controlled independently in both directions. These data offer new insights into the physiological behavior of in vivo cohesin-mediated loop extrusion and further our understanding of the principles that underlie genome organization.


Subject(s)
Chromatin , Chromosomal Proteins, Non-Histone , Animals , Chromatin/genetics , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Polymers/metabolism , Mammals/metabolism , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL