Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.664
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 93(1): 21-46, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38594943

ABSTRACT

DNA replication and transcription occur in all living cells across all domains of life. Both essential processes occur simultaneously on the same template, leading to conflicts between the macromolecular machines that perform these functions. Numerous studies over the past few decades demonstrate that this is an inevitable problem in both prokaryotic and eukaryotic cells. We have learned that conflicts lead to replication fork reversal, breaks in the DNA, R-loop formation, topological stress, and mutagenesis and can ultimately impact evolution. Recent studies have also provided insight into the various mechanisms that mitigate, resolve, and allow tolerance of conflicts and how conflicts result in pathological consequences across divergent species. In this review, we summarize our current knowledge regarding the outcomes of the encounters between replication and transcription machineries and explore how these clashes are dealt with across species.


Subject(s)
DNA Replication , Transcription, Genetic , Humans , Animals , Chromosomes/metabolism , Chromosomes/genetics , Chromosomes/chemistry , R-Loop Structures , DNA/metabolism , DNA/genetics , DNA/chemistry
2.
Annu Rev Biochem ; 92: 15-41, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37137166

ABSTRACT

SMC (structural maintenance of chromosomes) protein complexes are an evolutionarily conserved family of motor proteins that hold sister chromatids together and fold genomes throughout the cell cycle by DNA loop extrusion. These complexes play a key role in a variety of functions in the packaging and regulation of chromosomes, and they have been intensely studied in recent years. Despite their importance, the detailed molecular mechanism for DNA loop extrusion by SMC complexes remains unresolved. Here, we describe the roles of SMCs in chromosome biology and particularly review in vitro single-molecule studies that have recently advanced our understanding of SMC proteins. We describe the mechanistic biophysical aspects of loop extrusion that govern genome organization and its consequences.


Subject(s)
Chromosomal Proteins, Non-Histone , Multiprotein Complexes , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Multiprotein Complexes/chemistry , Chromosomes/genetics , Chromosomes/metabolism , DNA/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
3.
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
4.
Cell ; 184(11): 2860-2877.e22, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33964210

ABSTRACT

Most human embryos are aneuploid. Aneuploidy frequently arises during the early mitotic divisions of the embryo, but its origin remains elusive. Human zygotes that cluster their nucleoli at the pronuclear interface are thought to be more likely to develop into healthy euploid embryos. Here, we show that the parental genomes cluster with nucleoli in each pronucleus within human and bovine zygotes, and clustering is required for the reliable unification of the parental genomes after fertilization. During migration of intact pronuclei, the parental genomes polarize toward each other in a process driven by centrosomes, dynein, microtubules, and nuclear pore complexes. The maternal and paternal chromosomes eventually cluster at the pronuclear interface, in direct proximity to each other, yet separated. Parental genome clustering ensures the rapid unification of the parental genomes on nuclear envelope breakdown. However, clustering often fails, leading to chromosome segregation errors and micronuclei, incompatible with healthy embryo development.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Development/genetics , Aneuploidy , Animals , Cattle , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Centrosome/metabolism , Chromosome Segregation/physiology , Chromosomes/metabolism , Fertilization/genetics , Humans , Male , Microtubules/metabolism , Mitosis , Oocytes/metabolism , Spermatozoa/metabolism , Zygote/metabolism
5.
Cell ; 184(23): 5775-5790.e30, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34739832

ABSTRACT

RNA, DNA, and protein molecules are highly organized within three-dimensional (3D) structures in the nucleus. Although RNA has been proposed to play a role in nuclear organization, exploring this has been challenging because existing methods cannot measure higher-order RNA and DNA contacts within 3D structures. To address this, we developed RNA & DNA SPRITE (RD-SPRITE) to comprehensively map the spatial organization of RNA and DNA. These maps reveal higher-order RNA-chromatin structures associated with three major classes of nuclear function: RNA processing, heterochromatin assembly, and gene regulation. These data demonstrate that hundreds of ncRNAs form high-concentration territories throughout the nucleus, that specific RNAs are required to recruit various regulators into these territories, and that these RNAs can shape long-range DNA contacts, heterochromatin assembly, and gene expression. These results demonstrate a mechanism where RNAs form high-concentration territories, bind to diffusible regulators, and guide them into compartments to regulate essential nuclear functions.


Subject(s)
Cell Nucleus/metabolism , RNA/metabolism , Animals , Cell Nucleus/drug effects , Chromobox Protein Homolog 5/metabolism , Chromosomes/metabolism , DNA/metabolism , DNA, Satellite/metabolism , DNA-Binding Proteins/metabolism , Dactinomycin/pharmacology , Female , Genome , HEK293 Cells , Heterochromatin/metabolism , Humans , Mice , Models, Biological , Multigene Family , RNA Polymerase II/metabolism , RNA Processing, Post-Transcriptional/drug effects , RNA Processing, Post-Transcriptional/genetics , RNA Splicing/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA-Binding Proteins/metabolism , Transcription, Genetic/drug effects
6.
Nat Rev Mol Cell Biol ; 24(9): 633-650, 2023 09.
Article in English | MEDLINE | ID: mdl-37231112

ABSTRACT

Many cellular processes require large-scale rearrangements of chromatin structure. Structural maintenance of chromosomes (SMC) protein complexes are molecular machines that can provide structure to chromatin. These complexes can connect DNA elements in cis, walk along DNA, build and processively enlarge DNA loops and connect DNA molecules in trans to hold together the sister chromatids. These DNA-shaping abilities place SMC complexes at the heart of many DNA-based processes, including chromosome segregation in mitosis, transcription control and DNA replication, repair and recombination. In this Review, we discuss the latest insights into how SMC complexes such as cohesin, condensin and the SMC5-SMC6 complex shape DNA to direct these fundamental chromosomal processes. We also consider how SMC complexes, by building chromatin loops, can counteract the natural tendency of alike chromatin regions to cluster. SMC complexes thus control nuclear organization by participating in a molecular tug of war that determines the architecture of our genome.


Subject(s)
Chromatin , Chromosomes , Chromosomes/genetics , Chromosomes/metabolism , Chromatin/genetics , DNA/genetics , DNA Replication/genetics , Mitosis , Cell Cycle Proteins/chemistry
7.
Cell ; 182(6): 1381-1383, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32946780

ABSTRACT

Eukaryotic Argonaute proteins strictly mediate RNA-guided RNA interference. In contrast, prokaryotic Argonautes can utilize DNA guides to target complementary DNA sequences to protect their hosts against invading DNA. In this issue of Cell, Jolly and colleagues demonstrate that Thermus thermophilus Argonaute additionally participates in DNA replication by unlinking catenated chromosomes.


Subject(s)
Argonaute Proteins , Thermus thermophilus , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Chromosomes/metabolism , DNA Replication , Prokaryotic Cells/metabolism , Thermus thermophilus/genetics , Thermus thermophilus/metabolism
8.
Cell ; 182(6): 1474-1489.e23, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32841603

ABSTRACT

Widespread changes to DNA methylation and chromatin are well documented in cancer, but the fate of higher-order chromosomal structure remains obscure. Here we integrated topological maps for colon tumors and normal colons with epigenetic, transcriptional, and imaging data to characterize alterations to chromatin loops, topologically associated domains, and large-scale compartments. We found that spatial partitioning of the open and closed genome compartments is profoundly compromised in tumors. This reorganization is accompanied by compartment-specific hypomethylation and chromatin changes. Additionally, we identify a compartment at the interface between the canonical A and B compartments that is reorganized in tumors. Remarkably, similar shifts were evident in non-malignant cells that have accumulated excess divisions. Our analyses suggest that these topological changes repress stemness and invasion programs while inducing anti-tumor immunity genes and may therefore restrain malignant progression. Our findings call into question the conventional view that tumor-associated epigenomic alterations are primarily oncogenic.


Subject(s)
Chromatin/metabolism , Chromosomes/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Cell Division , Cellular Senescence/genetics , Chromatin Immunoprecipitation Sequencing , Chromosomes/genetics , Cohort Studies , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , DNA Methylation/genetics , Epigenomics , HCT116 Cells , Humans , In Situ Hybridization, Fluorescence , Microscopy, Electron, Transmission , Molecular Dynamics Simulation , RNA-Seq , Spatial Analysis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
Cell ; 182(6): 1545-1559.e18, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32846159

ABSTRACT

In many eukaryotes, Argonaute proteins, guided by short RNA sequences, defend cells against transposons and viruses. In the eubacterium Thermus thermophilus, the DNA-guided Argonaute TtAgo defends against transformation by DNA plasmids. Here, we report that TtAgo also participates in DNA replication. In vivo, TtAgo binds 15- to 18-nt DNA guides derived from the chromosomal region where replication terminates and associates with proteins known to act in DNA replication. When gyrase, the sole T. thermophilus type II topoisomerase, is inhibited, TtAgo allows the bacterium to finish replicating its circular genome. In contrast, loss of gyrase and TtAgo activity slows growth and produces long sausage-like filaments in which the individual bacteria are linked by DNA. Finally, wild-type T. thermophilus outcompetes an otherwise isogenic strain lacking TtAgo. We propose that the primary role of TtAgo is to help T. thermophilus disentangle the catenated circular chromosomes generated by DNA replication.


Subject(s)
Argonaute Proteins/metabolism , Bacterial Proteins/metabolism , DNA Gyrase/metabolism , DNA Replication/genetics , DNA/metabolism , Thermus thermophilus/metabolism , Argonaute Proteins/genetics , Bacterial Proteins/genetics , Cell Survival/drug effects , Cell Survival/genetics , Chromosomes/metabolism , Ciprofloxacin/pharmacology , DNA/genetics , DNA Replication/drug effects , Endonucleases/metabolism , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Models, Molecular , Recombinant Proteins , Recombination, Genetic/drug effects , Recombination, Genetic/genetics , Single Molecule Imaging , Tandem Mass Spectrometry , Thermus thermophilus/genetics , Thermus thermophilus/growth & development , Thermus thermophilus/ultrastructure , Topoisomerase II Inhibitors/pharmacology
10.
Cell ; 176(3): 520-534.e25, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30661750

ABSTRACT

Elucidating the global and local rules that govern genome-wide, hierarchical chromatin architecture remains a critical challenge. Current high-throughput chromosome conformation capture (Hi-C) technologies have identified large-scale chromatin structural motifs, such as topologically associating domains and looping. However, structural rules at the smallest or nucleosome scale remain poorly understood. Here, we coupled nucleosome-resolved Hi-C technology with simulated annealing-molecular dynamics (SA-MD) simulation to reveal 3D spatial distributions of nucleosomes and their genome-wide orientation in chromatin. Our method, called Hi-CO, revealed distinct nucleosome folding motifs across the yeast genome. Our results uncovered two types of basic secondary structural motifs in nucleosome folding: α-tetrahedron and ß-rhombus analogous to α helix and ß sheet motifs in protein folding. Using mutants and cell-cycle-synchronized cells, we further uncovered motifs with specific nucleosome positioning and orientation coupled to epigenetic features at individual loci. By illuminating molecular-level structure-function relationships in eukaryotic chromatin, our findings establish organizational principles of nucleosome folding.


Subject(s)
Chromatin/ultrastructure , Nucleosomes/ultrastructure , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Chromosomes/metabolism , Chromosomes/ultrastructure , Nucleosomes/genetics , Nucleosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Initiation Site
11.
Nat Rev Mol Cell Biol ; 22(4): 283-298, 2021 04.
Article in English | MEDLINE | ID: mdl-33564154

ABSTRACT

The regulation of telomere length in mammals is crucial for chromosome end-capping and thus for maintaining genome stability and cellular lifespan. This process requires coordination between telomeric protein complexes and the ribonucleoprotein telomerase, which extends the telomeric DNA. Telomeric proteins modulate telomere architecture, recruit telomerase to accessible telomeres and orchestrate the conversion of the newly synthesized telomeric single-stranded DNA tail into double-stranded DNA. Dysfunctional telomere maintenance leads to telomere shortening, which causes human diseases including bone marrow failure, premature ageing and cancer. Recent studies provide new insights into telomerase-related interactions (the 'telomere replisome') and reveal new challenges for future telomere structural biology endeavours owing to the dynamic nature of telomere architecture and the great number of structures that telomeres form. In this Review, we discuss recently determined structures of the shelterin and CTC1-STN1-TEN1 (CST) complexes, how they may participate in the regulation of telomere replication and chromosome end-capping, and how disease-causing mutations in their encoding genes may affect specific functions. Major outstanding questions in the field include how all of the telomere components assemble relative to each other and how the switching between different telomere structures is achieved.


Subject(s)
Chromatin/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Animals , Chromosomes/metabolism , DNA/metabolism , Humans , Telomerase/metabolism
12.
Nat Rev Mol Cell Biol ; 22(8): 511-528, 2021 08.
Article in English | MEDLINE | ID: mdl-33953379

ABSTRACT

Understanding how chromatin is folded in the nucleus is fundamental to understanding its function. Although 3D genome organization has been historically difficult to study owing to a lack of relevant methodologies, major technological breakthroughs in genome-wide mapping of chromatin contacts and advances in imaging technologies in the twenty-first century considerably improved our understanding of chromosome conformation and nuclear architecture. In this Review, we discuss methods of 3D genome organization analysis, including sequencing-based techniques, such as Hi-C and its derivatives, Micro-C, DamID and others; microscopy-based techniques, such as super-resolution imaging coupled with fluorescence in situ hybridization (FISH), multiplex FISH, in situ genome sequencing and live microscopy methods; and computational and modelling approaches. We describe the most commonly used techniques and their contribution to our current knowledge of nuclear architecture and, finally, we provide a perspective on up-and-coming methods that open possibilities for future major discoveries.


Subject(s)
Chromatin/chemistry , Genome , Chromatin/genetics , Chromatin/metabolism , Chromosome Mapping , Chromosomes/chemistry , Chromosomes/genetics , Chromosomes/metabolism , Computational Biology , High-Throughput Nucleotide Sequencing , Humans , Microscopy , Models, Molecular , Sequence Analysis, DNA
13.
Cell ; 174(2): 363-376.e16, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887381

ABSTRACT

Visualization of the transcriptome and the nuclear organization in situ has been challenging for single-cell analysis. Here, we demonstrate a multiplexed single-molecule in situ method, intron seqFISH, that allows imaging of 10,421 genes at their nascent transcription active sites in single cells, followed by mRNA and lncRNA seqFISH and immunofluorescence. This nascent transcriptome-profiling method can identify different cell types and states with mouse embryonic stem cells and fibroblasts. The nascent sites of RNA synthesis tend to be localized on the surfaces of chromosome territories, and their organization in individual cells is highly variable. Surprisingly, the global nascent transcription oscillated asynchronously in individual cells with a period of 2 hr in mouse embryonic stem cells, as well as in fibroblasts. Together, spatial genomics of the nascent transcriptome by intron seqFISH reveals nuclear organizational principles and fast dynamics in single cells that are otherwise obscured.


Subject(s)
In Situ Hybridization, Fluorescence/methods , Transcriptome , Animals , Catalytic Domain , Cell Line , Chromosomes/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Introns , Mice , Microscopy, Fluorescence , Microscopy, Video , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Single-Cell Analysis
14.
Cell ; 173(5): 1165-1178.e20, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29706548

ABSTRACT

Cohesin extrusion is thought to play a central role in establishing the architecture of mammalian genomes. However, extrusion has not been visualized in vivo, and thus, its functional impact and energetics are unknown. Using ultra-deep Hi-C, we show that loop domains form by a process that requires cohesin ATPases. Once formed, however, loops and compartments are maintained for hours without energy input. Strikingly, without ATP, we observe the emergence of hundreds of CTCF-independent loops that link regulatory DNA. We also identify architectural "stripes," where a loop anchor interacts with entire domains at high frequency. Stripes often tether super-enhancers to cognate promoters, and in B cells, they facilitate Igh transcription and recombination. Stripe anchors represent major hotspots for topoisomerase-mediated lesions, which promote chromosomal translocations and cancer. In plasmacytomas, stripes can deregulate Igh-translocated oncogenes. We propose that higher organisms have coopted cohesin extrusion to enhance transcription and recombination, with implications for tumor development.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Genome , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomes/metabolism , DNA-Binding Proteins , Humans , Mice , Mutagenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Cohesins
15.
Cell ; 169(7): 1214-1227.e18, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28622508

ABSTRACT

Higher eukaryotic chromosomes are organized into topologically constrained functional domains; however, the molecular mechanisms required to sustain these complex interphase chromatin structures are unknown. A stable matrix underpinning nuclear organization was hypothesized, but the idea was abandoned as more dynamic models of chromatin behavior became prevalent. Here, we report that scaffold attachment factor A (SAF-A), originally identified as a structural nuclear protein, interacts with chromatin-associated RNAs (caRNAs) via its RGG domain to regulate human interphase chromatin structures in a transcription-dependent manner. Mechanistically, this is dependent on SAF-A's AAA+ ATPase domain, which mediates cycles of protein oligomerization with caRNAs, in response to ATP binding and hydrolysis. SAF-A oligomerization decompacts large-scale chromatin structure while SAF-A loss or monomerization promotes aberrant chromosome folding and accumulation of genome damage. Our results show that SAF-A and caRNAs form a dynamic, transcriptionally responsive chromatin mesh that organizes large-scale chromosome structures and protects the genome from instability.


Subject(s)
Chromosomes/metabolism , Genomic Instability , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , RNA, Small Nuclear/metabolism , Amino Acid Sequence , Chromatin , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein U/chemistry , Humans , Interphase , Models, Molecular , Sequence Alignment , Transcription, Genetic
16.
Cell ; 170(4): 760-773.e15, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28781165

ABSTRACT

Inaccurate repair of broken chromosomes generates structural variants that can fuel evolution and inflict pathology. We describe a novel rearrangement mechanism in which translocation between intact chromosomes is induced by a lesion on a third chromosome. This multi-invasion-induced rearrangement (MIR) stems from a homologous recombination byproduct, where a broken DNA end simultaneously invades two intact donors. No homology is required between the donors, and the intervening sequence from the invading molecule is inserted at the translocation site. MIR is stimulated by increasing homology length and spatial proximity of the donors and depends on the overlapping activities of the structure-selective endonucleases Mus81-Mms4, Slx1-Slx4, and Yen1. Conversely, the 3'-flap nuclease Rad1-Rad10 and enzymes known to disrupt recombination intermediates (Sgs1-Top3-Rmi1, Srs2, and Mph1) inhibit MIR. Resolution of MIR intermediates propagates secondary chromosome breaks that frequently cause additional rearrangements. MIR features have implications for the formation of simple and complex rearrangements underlying human pathologies.


Subject(s)
Chromosomes/metabolism , DNA Repair , Genomic Instability , Translocation, Genetic , DNA Breaks, Double-Stranded , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Homologous Recombination , Humans , Saccharomyces cerevisiae/genetics
17.
Cell ; 171(2): 305-320.e24, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985562

ABSTRACT

The human genome folds to create thousands of intervals, called "contact domains," that exhibit enhanced contact frequency within themselves. "Loop domains" form because of tethering between two loci-almost always bound by CTCF and cohesin-lying on the same chromosome. "Compartment domains" form when genomic intervals with similar histone marks co-segregate. Here, we explore the effects of degrading cohesin. All loop domains are eliminated, but neither compartment domains nor histone marks are affected. Loss of loop domains does not lead to widespread ectopic gene activation but does affect a significant minority of active genes. In particular, cohesin loss causes superenhancers to co-localize, forming hundreds of links within and across chromosomes and affecting the regulation of nearby genes. We then restore cohesin and monitor the re-formation of each loop. Although re-formation rates vary greatly, many megabase-sized loops recovered in under an hour, consistent with a model where loop extrusion is rapid.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Genome, Human , Repressor Proteins/metabolism , CCCTC-Binding Factor , Cell Line, Tumor , DNA-Binding Proteins , Enhancer Elements, Genetic , Histone Code , Humans , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Phosphoproteins/metabolism , Cohesins
18.
Cell ; 171(3): 588-600.e24, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28988770

ABSTRACT

Condensin protein complexes coordinate the formation of mitotic chromosomes and thereby ensure the successful segregation of replicated genomes. Insights into how condensin complexes bind to chromosomes and alter their topology are essential for understanding the molecular principles behind the large-scale chromatin rearrangements that take place during cell divisions. Here, we identify a direct DNA-binding site in the eukaryotic condensin complex, which is formed by its Ycg1Cnd3 HEAT-repeat and Brn1Cnd2 kleisin subunits. DNA co-crystal structures reveal a conserved, positively charged groove that accommodates the DNA double helix. A peptide loop of the kleisin subunit encircles the bound DNA and, like a safety belt, prevents its dissociation. Firm closure of the kleisin loop around DNA is essential for the association of condensin complexes with chromosomes and their DNA-stimulated ATPase activity. Our data suggest a sophisticated molecular basis for anchoring condensin complexes to chromosomes that enables the formation of large-sized chromatin loops.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomes/metabolism , DNA-Binding Proteins/metabolism , Eukaryota/metabolism , Fungal Proteins/metabolism , Multiprotein Complexes/metabolism , Adenosine Triphosphatases/chemistry , Amino Acid Sequence , Chaetomium/metabolism , Chromosomes/chemistry , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , Eukaryota/chemistry , Fungal Proteins/chemistry , HeLa Cells , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Saccharomyces cerevisiae/metabolism , Sequence Alignment
19.
Mol Cell ; 84(2): 186-188, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38242096

ABSTRACT

Two recent studies in Molecular Cell1 and Nature2 show that evicted RNA polymerases reassociate rapidly with post-replicative chromatin and proceed into an unusual transcription cycle, bypassing regular controls and creating a temporary window for altered gene expression.


Subject(s)
Chromatin , DNA Replication , Chromatin/genetics , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Chromosomes/metabolism , Transcription, Genetic
20.
Mol Cell ; 84(5): 814-815, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38458170

ABSTRACT

In this issue of Molecular Cell, two papers provide insight into atypical structural maintenance of chromosomes protein complexes (SMCs). Jeppsson et al.1 link Smc5/6 to supercoiled DNA, and Roisné-Hamelin et al.2 show how Wadjet SMC bends and cleaves invading DNAs.


Subject(s)
Cell Cycle Proteins , Chromosomes , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomes/metabolism , DNA , DNA Repair , DNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL