Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.566
Filter
1.
Methods Mol Biol ; 2856: 445-453, 2025.
Article in English | MEDLINE | ID: mdl-39283468

ABSTRACT

Cohesin is a protein complex that plays a key role in regulating chromosome structure and gene expression. While next-generation sequencing technologies have provided extensive information on various aspects of cohesin, integrating and exploring the vast datasets associated with cohesin are not straightforward. CohesinDB ( https://cohesindb.iqb.u-tokyo.ac.jp ) offers a web-based interface for browsing, searching, analyzing, visualizing, and downloading comprehensive multiomics cohesin information in human cells. In this protocol, we introduce how to utilize CohesinDB to facilitate research on transcriptional regulation and chromatin organization.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , Web Browser , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Humans , Software , Computational Biology/methods , Genomics/methods , Databases, Genetic , Chromatin/metabolism , Chromatin/genetics , Internet , Multiomics
2.
Methods Mol Biol ; 2856: 11-22, 2025.
Article in English | MEDLINE | ID: mdl-39283444

ABSTRACT

The Structural Maintenance of Chromosomes (SMC) protein complexes are DNA-binding molecular machines required to shape chromosomes into functional units and to safeguard the genome through cell division. These ring-shaped multi-subunit protein complexes, which are present in all kingdoms of life, achieve this by organizing chromosomes in three-dimensional space. Mechanistically, the SMC complexes hydrolyze ATP to either stably entrap DNA molecules within their lumen, or rapidly reel DNA into large loops, which allow them to link two stretches of DNA in cis or trans. In this chapter, the canonical structure of the SMC complexes is first introduced, followed by a description of the composition and general functions of the main types of eukaryotic and prokaryotic SMC complexes. Thereafter, the current model for how SMC complexes perform in vitro DNA loop extrusion is presented. Lastly, chromosome loop formation by SMC complexes is introduced, and how the DNA loop extrusion mechanism contributes to chromosome looping by SMC complexes in cells is discussed.


Subject(s)
Chromosomes , Chromosomes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/chemistry , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA/chemistry , DNA/metabolism , DNA/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Adenosine Triphosphate/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry
3.
Methods Mol Biol ; 2856: 71-78, 2025.
Article in English | MEDLINE | ID: mdl-39283447

ABSTRACT

Hi-C reads, which represent ligation events between different regions of the genome, must be processed into matrices of interaction frequencies for downstream analysis. Here, I describe a procedure for mapping Hi-C reads to the genome and conversion of mapped reads into the HOMER tag directory format and interaction matrix format for visualization with Juicebox. The method is demonstrated for the mouse composite X chromosome in which reads from the active and inactive X chromosomes are combined after mock DMSO treatment or targeted degradation of cohesin.


Subject(s)
X Chromosome , Animals , X Chromosome/genetics , Mice , Software , Cohesins , Chromosome Mapping/methods , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Computational Biology/methods
4.
Science ; 385(6713): 1091-1097, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39236163

ABSTRACT

The centromere, a chromosome locus defined by the histone H3-like protein centromeric protein A (CENP-A), promotes assembly of the kinetochore to bind microtubules during cell division. Centromere maintenance requires CENP-A to be actively replenished by dedicated protein machinery in the early G1 phase of the cell cycle to compensate for its dilution after DNA replication. Cyclin-dependent kinases (CDKs) limit CENP-A deposition to once per cell cycle and function as negative regulators outside of early G1. Antithetically, Polo-like kinase 1 (PLK1) promotes CENP-A deposition in early G1, but the molecular details of this process are still unknown. We reveal here a phosphorylation network that recruits PLK1 to the deposition machinery to control a conformational switch required for licensing the CENP-A deposition reaction. Our findings clarify how PLK1 contributes to the epigenetic maintenance of centromeres.


Subject(s)
Cell Cycle Proteins , Centromere Protein A , Centromere , Chromosomal Proteins, Non-Histone , Epigenesis, Genetic , Polo-Like Kinase 1 , Humans , Cell Cycle Proteins/metabolism , Centromere/metabolism , Centromere Protein A/metabolism , Chromosomal Proteins, Non-Histone/metabolism , G1 Phase , HeLa Cells , Kinetochores/metabolism , Phosphorylation , Polo-Like Kinase 1/genetics , Polo-Like Kinase 1/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics
5.
J Exp Med ; 221(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39297882

ABSTRACT

The establishment of long-lasting immunity against pathogens is facilitated by the germinal center (GC) reaction, during which B cells increase their antibody affinity and differentiate into antibody-secreting cells (ASC) and memory cells. These events involve modifications in chromatin packaging that orchestrate the profound restructuring of gene expression networks that determine cell fate. While several chromatin remodelers were implicated in lymphocyte functions, less is known about SMARCA5. Here, using ribosomal pull-down for analyzing translated genes in GC B cells, coupled with functional experiments in mice, we identified SMARCA5 as a key chromatin remodeler in B cells. While the naive B cell compartment remained unaffected following conditional depletion of Smarca5, effective proliferation during B cell activation, immunoglobulin class switching, and as a result GC formation and ASC differentiation were impaired. Single-cell multiomic sequencing analyses revealed that SMARCA5 is crucial for facilitating the transcriptional modifications and genomic accessibility of genes that support B cell activation and differentiation. These findings offer novel insights into the functions of SMARCA5, which can be targeted in various human pathologies.


Subject(s)
B-Lymphocytes , Cell Differentiation , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Germinal Center , Animals , Germinal Center/immunology , Germinal Center/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Mice , Mice, Inbred C57BL , Lymphocyte Activation/immunology , Immunoglobulin Class Switching/genetics , Adenosine Triphosphatases
6.
Nat Commun ; 15(1): 7805, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242537

ABSTRACT

Beyond its essential roles in ensuring faithful chromosome segregation and genomic stability, the human Smc5/6 complex acts as an antiviral factor. It binds to and impedes the transcription of extrachromosomal DNA templates; an ability which is lost upon integration of the DNA into the chromosome. How the complex distinguishes among different DNA templates is unknown. Here we show that, in human cells, Smc5/6 preferentially binds to circular rather than linear extrachromosomal DNA. We further demonstrate that the transcriptional process, per se, and particularly the accumulation of DNA secondary structures known to be substrates for topoisomerases, is responsible for Smc5/6 recruitment. More specifically, we find that in vivo Smc5/6 binds to positively supercoiled DNA. Those findings, in conjunction with our genome-wide Smc5/6 binding analysis showing that Smc5/6 localizes at few but highly transcribed chromosome loci, not only unveil a previously unforeseen role of Smc5/6 in DNA topology management during transcription but highlight the significance of sensing DNA topology as an antiviral defense mechanism.


Subject(s)
Cell Cycle Proteins , DNA, Superhelical , Transcription, Genetic , Humans , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA, Superhelical/metabolism , DNA, Superhelical/genetics , Protein Binding , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA/metabolism , DNA/genetics , Nucleic Acid Conformation , DNA, Circular/metabolism , DNA, Circular/genetics
7.
Proc Natl Acad Sci U S A ; 121(38): e2402518121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39254997

ABSTRACT

The in vivo three-dimensional genomic architecture of adult mature neurons at homeostasis and after medically relevant perturbations such as axonal injury remains elusive. Here, we address this knowledge gap by mapping the three-dimensional chromatin architecture and gene expression program at homeostasis and after sciatic nerve injury in wild-type and cohesin-deficient mouse sensory dorsal root ganglia neurons via combinatorial Hi-C, promoter-capture Hi-C, CUT&Tag for H3K27ac and RNA-seq. We find that genes involved in axonal regeneration form long-range, complex chromatin loops, and that cohesin is required for the full induction of the regenerative transcriptional program. Importantly, loss of cohesin results in disruption of chromatin architecture and severely impaired nerve regeneration. Complex enhancer-promoter loops are also enriched in the human fetal cortical plate, where the axonal growth potential is highest, and are lost in mature adult neurons. Together, these data provide an original three-dimensional chromatin map of adult sensory neurons in vivo and demonstrate a role for cohesin-dependent long-range promoter interactions in nerve regeneration.


Subject(s)
Axons , Chromatin , Cohesins , Nerve Regeneration , Promoter Regions, Genetic , Sensory Receptor Cells , Animals , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Mice , Promoter Regions, Genetic/genetics , Chromatin/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Axons/metabolism , Axons/physiology , Humans , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Enhancer Elements, Genetic/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Sciatic Nerve/metabolism
8.
Nature ; 633(8031): 941-951, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39294374

ABSTRACT

Subcellular protein localization regulates protein function and can be corrupted in cancers1 and neurodegenerative diseases2,3. The rewiring of localization to address disease-driving phenotypes would be an attractive targeted therapeutic approach. Molecules that harness the trafficking of a shuttle protein to control the subcellular localization of a target protein could enforce targeted protein relocalization and rewire the interactome. Here we identify a collection of shuttle proteins with potent ligands amenable to incorporation into targeted relocalization-activating molecules (TRAMs), and use these to relocalize endogenous proteins. Using a custom imaging analysis pipeline, we show that protein steady-state localization can be modulated through molecular coupling to shuttle proteins containing sufficiently strong localization sequences and expressed in the necessary abundance. We analyse the TRAM-induced relocalization of different proteins and then use nuclear hormone receptors as shuttles to redistribute disease-driving mutant proteins such as SMARCB1Q318X, TDP43ΔNLS and FUSR495X. TRAM-mediated relocalization of FUSR495X to the nucleus from the cytoplasm correlated with a reduction in the number of stress granules in a model of cellular stress. With methionyl aminopeptidase 2 and poly(ADP-ribose) polymerase 1 as endogenous cytoplasmic and nuclear shuttles, respectively, we demonstrate relocalization of endogenous PRMT9, SOS1 and FKBP12. Small-molecule-mediated redistribution of nicotinamide nucleotide adenylyltransferase 1 from nuclei to axons in primary neurons was able to slow axonal degeneration and pharmacologically mimic the genetic WldS gain-of-function phenotype in mice resistant to certain types of neurodegeneration4. The concept of targeted protein relocalization could therefore inspire approaches for treating disease through interactome rewiring.


Subject(s)
Protein Transport , Humans , Animals , Mice , Cell Nucleus/metabolism , Ligands , Chromosomal Proteins, Non-Histone/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Female , Male , Neurons/metabolism
9.
Science ; 385(6713): 1098-1104, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39236175

ABSTRACT

Accurate chromosome segregation requires the attachment of microtubules to centromeres, epigenetically defined by the enrichment of CENP-A nucleosomes. During DNA replication, CENP-A nucleosomes undergo dilution. To preserve centromere identity, correct amounts of CENP-A must be restored in a cell cycle-controlled manner orchestrated by the Mis18 complex (Mis18α-Mis18ß-Mis18BP1). We demonstrate here that PLK1 interacts with the Mis18 complex by recognizing self-primed phosphorylations of Mis18α (Ser54) and Mis18BP1 (Thr78 and Ser93) through its Polo-box domain. Disrupting these phosphorylations perturbed both centromere recruitment of the CENP-A chaperone HJURP and new CENP-A loading. Biochemical and functional analyses showed that phosphorylation of Mis18α and PLK1 binding were required to activate Mis18α-Mis18ß and promote Mis18 complex-HJURP interaction. Thus, our study reveals key molecular events underpinning the licensing role of PLK1 in ensuring accurate centromere inheritance.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Centromere Protein A , Centromere , Chromosomal Proteins, Non-Histone , Polo-Like Kinase 1 , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Humans , Cell Cycle Proteins/metabolism , Centromere/metabolism , Centromere Protein A/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , DNA-Binding Proteins/metabolism , HeLa Cells , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism
10.
Nat Commun ; 15(1): 7646, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223123

ABSTRACT

Despite their prevalent cancer implications, the in vivo dynamics of SWI/SNF chromatin remodelers and how misregulation of such dynamics underpins cancer remain poorly understood. Using live-cell single-molecule tracking, we quantify the intranuclear diffusion and chromatin-binding of three key subunits common to all major human SWI/SNF remodeler complexes (BAF57, BAF155 and BRG1), and resolve two temporally distinct stable binding modes for the fully assembled complex. Super-resolved density mapping reveals heterogeneous, nanoscale remodeler binding "hotspots" across the nucleoplasm where multiple binding events (especially longer-lived ones) preferentially cluster. Importantly, we uncover distinct roles of the bromodomain in modulating chromatin binding/targeting in a DNA-accessibility-dependent manner, pointing to a model where successive longer-lived binding within "hotspots" leads to sustained productive remodeling. Finally, systematic comparison of six common BRG1 mutants implicated in various cancers unveils alterations in chromatin-binding dynamics unique to each mutant, shedding insight into a multi-modal landscape regulating the spatio-temporal organizational dynamics of SWI/SNF remodelers.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin , Chromosomal Proteins, Non-Histone , DNA Helicases , Neoplasms , Nuclear Proteins , Single Molecule Imaging , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Single Molecule Imaging/methods , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , DNA/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Protein Binding , Mutation , Cell Line, Tumor , Protein Domains , Adenosine Triphosphatases
11.
Mol Cell ; 84(17): 3167-3169, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39241749

ABSTRACT

In this issue of Molecular Cell, Sahu et al.1 find that shielding heterochromatin from SWI/SNF chromatin remodelers is essential to maintain and epigenetically propagate pre-existing heterochromatin domains, whereas SWI/SNF action protects facultative heterochromatic regions from premature silencing.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Heterochromatin , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Epigenesis, Genetic , Gene Silencing , Heterochromatin/metabolism , Heterochromatin/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
12.
Curr Genet ; 70(1): 15, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39235627

ABSTRACT

Chromatin remodelling complexes (CRC) are ATP-dependent molecular machines important for the dynamic organization of nucleosomes along eukaryotic DNA. CRCs SWI/SNF, RSC and INO80 can move positioned nucleosomes in promoter DNA, leading to nucleosome-depleted regions which facilitate access of general transcription factors. This function is strongly supported by transcriptional activators being able to interact with subunits of various CRCs. In this work we show that SWI/SNF subunits Swi1, Swi2, Snf5 and Snf6 can bind to activation domains of Ino2 required for expression of phospholipid biosynthetic genes in yeast. We identify an activator binding domain (ABD) of ATPase Swi2 and show that this ABD is functionally dispensable, presumably because ABDs of other SWI/SNF subunits can compensate for the loss. In contrast, mutational characterization of the ABD of the Swi2-related ATPase Sth1 revealed that some conserved basic and hydrophobic amino acids within this domain are essential for the function of Sth1. While ABDs of Swi2 and Sth1 define separate functional protein domains, mapping of an ABD within ATPase Ino80 showed co-localization with its HSA domain also required for binding actin-related proteins. Comparative interaction studies finally demonstrated that several unrelated activators each exhibit a specific binding pattern with ABDs of Swi2, Sth1 and Ino80.


Subject(s)
Adenosine Triphosphatases , Chromatin Assembly and Disassembly , DNA-Binding Proteins , Protein Binding , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Transcriptional Activation , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Fungal , Protein Domains , Nuclear Proteins , Cell Cycle Proteins , Basic Helix-Loop-Helix Transcription Factors
14.
Mol Cell ; 84(17): 3237-3253.e6, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39178861

ABSTRACT

Homology search is a central step of DNA double-strand break (DSB) repair by homologous recombination (HR). How it operates in cells remains elusive. We developed a Hi-C-based methodology to map single-stranded DNA (ssDNA) contacts genome-wide in S. cerevisiae, which revealed two main homology search phases. Initial search conducted by short Rad51-ssDNA nucleoprotein filaments (NPFs) is confined in cis by cohesin-mediated chromatin loop folding. Progressive growth of stiff NPFs enables exploration of distant genomic sites. Long-range resection drives this transition from local to genome-wide search by increasing the probability of assembling extensive NPFs. DSB end-tethering promotes coordinated search by opposite NPFs. Finally, an autonomous genetic element on chromosome III engages the NPF, which stimulates homology search in its vicinity. This work reveals the mechanism of the progressive expansion of homology search that is orchestrated by chromatin organizers, long-range resection, end-tethering, and specialized genetic elements and that exploits the stiff NPF structure conferred by Rad51 oligomerization.


Subject(s)
DNA Breaks, Double-Stranded , DNA, Fungal , DNA, Single-Stranded , Rad51 Recombinase , Recombinational DNA Repair , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Chromatin/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cohesins
16.
DNA Repair (Amst) ; 142: 103742, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39137555

ABSTRACT

At the core of cellular life lies a carefully orchestrated interplay of DNA replication, recombination, chromatin assembly, sister-chromatid cohesion and transcription. These fundamental processes, while seemingly discrete, are inextricably linked during genome replication. A set of replisome factors integrate various DNA transactions and contribute to the transient formation of sister chromatid junctions involving either the cohesin complex or DNA four-way junctions. The latter structures serve DNA damage bypass and may have additional roles in replication fork stabilization or in marking regions of replication fork blockage. Here, we will discuss these concepts based on the ability of one replisome component, Ctf4, to act as a hub and functionally link these processes during DNA replication to ensure genome maintenance.


Subject(s)
DNA Replication , DNA-Binding Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae Proteins/metabolism , DNA-Binding Proteins/metabolism , Genomic Instability , Cell Cycle Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , DNA Damage , Chromosomal Proteins, Non-Histone/metabolism , Cohesins
17.
EMBO Rep ; 25(9): 4062-4077, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39179892

ABSTRACT

Acute protein knockdown is a powerful approach to dissecting protein function in dynamic cellular processes. We previously reported an improved auxin-inducible degron system, AID2, but recently noted that its ability to induce degradation of some essential replication factors, such as ORC1 and CDC6, was not enough to induce lethality. Here, we present combinational degron technologies to control two proteins or enhance target depletion. For this purpose, we initially compare PROTAC-based degrons, dTAG and BromoTag, with AID2 to reveal their key features and then demonstrate control of cohesin and condensin with AID2 and BromoTag, respectively. We develop a double-degron system with AID2 and BromoTag to enhance target depletion and accelerate depletion kinetics and demonstrate that both ORC1 and CDC6 are pivotal for MCM loading. Finally, we show that co-depletion of ORC1 and CDC6 by the double-degron system completely suppresses DNA replication, and the cells enter mitosis with single-chromatid chromosomes, indicating that DNA replication is uncoupled from cell cycle control. Our combinational degron technologies will expand the application scope for functional analyses.


Subject(s)
Adenosine Triphosphatases , Cell Cycle Proteins , DNA Replication , DNA-Binding Proteins , Multiprotein Complexes , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Humans , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Multiprotein Complexes/metabolism , Origin Recognition Complex/metabolism , Origin Recognition Complex/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Gene Knockdown Techniques , Cohesins , Mitosis/drug effects , Mitosis/genetics , Proteolysis , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , Minichromosome Maintenance Proteins/genetics , Degrons
18.
Bioessays ; 46(10): e2400137, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39093600

ABSTRACT

TAD boundaries are genomic elements that separate biological processes in neighboring domains by blocking DNA loops that are formed through Cohesin-mediated loop extrusion. Most TAD boundaries consist of arrays of binding sites for the CTCF protein, whose interaction with the Cohesin complex blocks loop extrusion. TAD boundaries are not fully impermeable though and allow a limited amount of inter-TAD loop formation. Based on the reanalysis of Nano-C data, a multicontact Chromosome Conformation Capture assay, we propose a model whereby clustered CTCF binding sites promote the successive stalling of Cohesin and subsequent dissociation from the chromatin. A fraction of Cohesin nonetheless achieves boundary read-through. Due to a constant rate of Cohesin dissociation elsewhere in the genome, the maximum length of inter-TAD loops is restricted though. We speculate that the DNA-encoded organization of stalling sites regulates TAD boundary permeability and discuss implications for enhancer-promoter loop formation and other genomic processes.


Subject(s)
CCCTC-Binding Factor , Cell Cycle Proteins , Chromatin , Chromosomal Proteins, Non-Histone , Cohesins , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Humans , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Chromatin/genetics , Animals , Binding Sites , Genome/genetics , Enhancer Elements, Genetic , Promoter Regions, Genetic/genetics , DNA/metabolism , DNA/genetics
19.
Plant J ; 120(1): 187-198, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39133829

ABSTRACT

Polycomb group (PcG) proteins are essential gene repressors in higher eukaryotes. However, how PcG proteins mediate transcriptional regulation of specific genes remains unknown. LIKE HETEROCHROMATIN PROTEIN 1 (LHP1), as a component of Polycomb Repression Complexes (PRC), epigenetically mediates several plant developmental processes together with PcG proteins. We observed physical interaction between MYB73 and LHP1 in vitro and in vivo. Genetic analysis indicated that myb73 mutants showed slightly late flowering, and the lhp1-3 myb73-2 double mutant exhibited delayed flowering and downregulated FT expression compared to lhp1-3. Chromatin immunoprecipitation and yeast one-hybrid assays revealed that MYB73 preferentially binds to the FT promoter. Additionally, our protoplast transient assays demonstrated that MYB73 activates to the FT promoter. Interestingly, the LHP1-MYB73 interaction is necessary to repress the FT promoter, suggesting that the LHP1-MYB73 interaction prevents FT activation by MYB73 in Arabidopsis. Our results show an example in which a chromatin regulator affects transcriptional regulation by negatively regulating a transcription factor through direct interaction.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Flowers , Gene Expression Regulation, Plant , Promoter Regions, Genetic , Transcription Factors , Transcriptional Activation , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/growth & development , Arabidopsis/physiology , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Flowers/genetics , Flowers/growth & development , Flowers/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Promoter Regions, Genetic/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Plants, Genetically Modified
20.
Mol Cell ; 84(18): 3406-3422.e6, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39173638

ABSTRACT

Partitioning of repressive from actively transcribed chromatin in mammalian cells fosters cell-type-specific gene expression patterns. While this partitioning is reconstructed during differentiation, the chromatin occupancy of the key insulator, CCCTC-binding factor (CTCF), is unchanged at the developmentally important Hox clusters. Thus, dynamic changes in chromatin boundaries must entail other activities. Given its requirement for chromatin loop formation, we examined cohesin-based chromatin occupancy without known insulators, CTCF and Myc-associated zinc-finger protein (MAZ), and identified a family of zinc-finger proteins (ZNFs), some of which exhibit tissue-specific expression. Two such ZNFs foster chromatin boundaries at the Hox clusters that are distinct from each other and from MAZ. PATZ1 was critical to the thoracolumbar boundary in differentiating motor neurons and mouse skeleton, while ZNF263 contributed to cervicothoracic boundaries. We propose that these insulating activities act with cohesin, alone or combinatorially, with or without CTCF, to implement precise positional identity and cell fate during development.


Subject(s)
CCCTC-Binding Factor , Cell Cycle Proteins , Chromatin , Chromosomal Proteins, Non-Histone , Cohesins , DNA-Binding Proteins , Animals , Chromatin/metabolism , Chromatin/genetics , Mice , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Transcription Factors/genetics , Motor Neurons/metabolism , Cell Differentiation , Zinc Fingers , Humans , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL