Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 569
Filter
1.
Cell ; 172(3): 465-477.e15, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29358048

ABSTRACT

The ring-shaped structural maintenance of chromosome (SMC) complexes are multi-subunit ATPases that topologically encircle DNA. SMC rings make vital contributions to numerous chromosomal functions, including mitotic chromosome condensation, sister chromatid cohesion, DNA repair, and transcriptional regulation. They are thought to do so by establishing interactions between more than one DNA. Here, we demonstrate DNA-DNA tethering by the purified fission yeast cohesin complex. DNA-bound cohesin efficiently and topologically captures a second DNA, but only if that is single-stranded DNA (ssDNA). Like initial double-stranded DNA (dsDNA) embrace, second ssDNA capture is ATP-dependent, and it strictly requires the cohesin loader complex. Second-ssDNA capture is relatively labile but is converted into stable dsDNA-dsDNA cohesion through DNA synthesis. Our study illustrates second-DNA capture by an SMC complex and provides a molecular model for the establishment of sister chromatid cohesion.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate/metabolism , Chromatids/metabolism , DNA Replication , Saccharomyces cerevisiae , Schizosaccharomyces , Cohesins
2.
Mol Cell ; 84(6): 1139-1148.e5, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38452765

ABSTRACT

Eukaryotic genomes are folded into DNA loops mediated by structural maintenance of chromosomes (SMC) complexes such as cohesin, condensin, and Smc5/6. This organization regulates different DNA-related processes along the cell cycle, such as transcription, recombination, segregation, and DNA repair. During the G2 stage, SMC-mediated DNA loops coexist with cohesin complexes involved in sister chromatid cohesion (SCC). However, the articulation between the establishment of SCC and the formation of SMC-mediated DNA loops along the chromatin remains unknown. Here, we show that SCC is indeed a barrier to cohesin-mediated DNA loop expansion along G2/M Saccharomyces cerevisiae chromosomes.


Subject(s)
Chromosomal Proteins, Non-Histone , Saccharomyces cerevisiae Proteins , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromatids/metabolism , Cohesins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA/genetics , DNA/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
3.
Mol Cell ; 83(17): 3049-3063.e6, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37591243

ABSTRACT

Cohesin connects CTCF-binding sites and other genomic loci in cis to form chromatin loops and replicated DNA molecules in trans to mediate sister chromatid cohesion. Whether cohesin uses distinct or related mechanisms to perform these functions is unknown. Here, we describe a cohesin hinge mutant that can extrude DNA into loops but is unable to mediate cohesion in human cells. Our results suggest that the latter defect arises during cohesion establishment. The observation that cohesin's cohesion and loop extrusion activities can be partially separated indicates that cohesin uses distinct mechanisms to perform these two functions. Unexpectedly, the same hinge mutant can also not be stopped by CTCF boundaries as well as wild-type cohesin. This suggests that cohesion establishment and cohesin's interaction with CTCF boundaries depend on related mechanisms and raises the possibility that both require transient hinge opening to entrap DNA inside the cohesin ring.


Subject(s)
Cell Cycle Proteins , Chromatids , Humans , Chromatids/genetics , Binding Sites , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Cohesins
4.
Genes Dev ; 35(19-20): 1368-1382, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34503989

ABSTRACT

The alternative PCNA loader containing CTF18-DCC1-CTF8 facilitates sister chromatid cohesion (SCC) by poorly defined mechanisms. Here we found that in DT40 cells, CTF18 acts complementarily with the Warsaw breakage syndrome DDX11 helicase in mediating SCC and proliferation. We uncover that the lethality and cohesion defects of ctf18 ddx11 mutants are associated with reduced levels of chromatin-bound cohesin and rescued by depletion of WAPL, a cohesin-removal factor. On the contrary, high levels of ESCO1/2 acetyltransferases that acetylate cohesin to establish SCC do not rescue ctf18 ddx11 phenotypes. Notably, the tight proximity of sister centromeres and increased anaphase bridges characteristic of WAPL-depleted cells are abrogated by loss of both CTF18 and DDX11 The results reveal that vertebrate CTF18 and DDX11 collaborate to provide sufficient amounts of chromatin-loaded cohesin available for SCC generation in the presence of WAPL-mediated cohesin-unloading activity. This process modulates chromosome structure and is essential for cellular proliferation in vertebrates.


Subject(s)
Chromatids , Chromosomal Proteins, Non-Histone , Animals , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromosomal Proteins, Non-Histone/genetics , Vertebrates/genetics , Cohesins
5.
EMBO J ; 43(12): 2424-2452, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714893

ABSTRACT

The 16-subunit Constitutive Centromere-associated Network (CCAN)-based inner kinetochore is well-known for connecting centromeric chromatin to the spindle-binding outer kinetochore. Here, we report a non-canonical role for the inner kinetochore in directly regulating sister-chromatid cohesion at centromeres. We provide biochemical, X-ray crystal structure, and intracellular ectopic localization evidence that the inner kinetochore directly binds cohesin, a ring-shaped multi-subunit complex that holds sister chromatids together from S-phase until anaphase onset. This interaction is mediated by binding of the 5-subunit CENP-OPQUR sub-complex of CCAN to the Scc1-SA2 sub-complex of cohesin. Mutation in the CENP-U subunit of the CENP-OPQUR complex that abolishes its binding to the composite interface between Scc1 and SA2 weakens centromeric cohesion, leading to premature separation of sister chromatids during delayed metaphase. We further show that CENP-U competes with the cohesin release factor Wapl for binding the interface of Scc1-SA2, and that the cohesion-protecting role for CENP-U can be bypassed by depleting Wapl. Taken together, this study reveals an inner kinetochore-bound pool of cohesin, which strengthens centromeric sister-chromatid cohesion to resist metaphase spindle pulling forces.


Subject(s)
Cell Cycle Proteins , Centromere , Chromatids , Chromosomal Proteins, Non-Histone , Kinetochores , Kinetochores/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Humans , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromatids/metabolism , Chromatids/genetics , Centromere/metabolism , Cohesins , HeLa Cells , Protein Binding , Crystallography, X-Ray
6.
EMBO J ; 43(19): 4173-4196, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39160275

ABSTRACT

The ring-shaped cohesin complex topologically entraps two DNA molecules to establish sister chromatid cohesion. Cohesin also shapes the interphase chromatin landscape with wide-ranging implications for gene regulation, and cohesin is thought to achieve this by actively extruding DNA loops without topologically entrapping DNA. The 'loop extrusion' hypothesis finds motivation from in vitro observations-whether this process underlies in vivo chromatin loop formation remains untested. Here, using the budding yeast S. cerevisiae, we generate cohesin variants that have lost their ability to extrude DNA loops but retain their ability to topologically entrap DNA. Analysis of these variants suggests that in vivo chromatin loops form independently of loop extrusion. Instead, we find that transcription promotes loop formation, and acts as an extrinsic motor that expands these loops and defines their ultimate positions. Our results necessitate a re-evaluation of the loop extrusion hypothesis. We propose that cohesin, akin to sister chromatid cohesion establishment at replication forks, forms chromatin loops by DNA-DNA capture at places of transcription, thus unifying cohesin's two roles in chromosome segregation and interphase genome organisation.


Subject(s)
Cell Cycle Proteins , Chromatin , Chromosomal Proteins, Non-Histone , Cohesins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/growth & development , Chromatin/metabolism , Chromatin/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , DNA, Fungal/metabolism , DNA, Fungal/genetics , Chromatids/metabolism , Chromatids/genetics , Transcription, Genetic
7.
Mol Cell ; 79(6): 902-916.e6, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32768407

ABSTRACT

A long-standing conundrum is how mitotic chromosomes can compact, as required for clean separation to daughter cells, while maintaining close parallel alignment of sister chromatids. Pursuit of this question, by high resolution 3D fluorescence imaging of living and fixed mammalian cells, has led to three discoveries. First, we show that the structural axes of separated sister chromatids are linked by evenly spaced "mini-axis" bridges. Second, when chromosomes first emerge as discrete units, at prophase, they are organized as co-oriented sister linear loop arrays emanating from a conjoined axis. We show that this same basic organization persists throughout mitosis, without helical coiling. Third, from prophase onward, chromosomes are deformed into sequential arrays of half-helical segments of alternating handedness (perversions), accompanied by correlated kinks. These arrays fluctuate dynamically over <15 s timescales. Together these discoveries redefine the foundation for thinking about the evolution of mitotic chromosomes as they prepare for anaphase segregation.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomes/genetics , DNA-Binding Proteins/genetics , Mitosis/genetics , Adenosine Triphosphatases/genetics , Anaphase/genetics , Animals , Cell Cycle Proteins/isolation & purification , Chromatids/genetics , Chromosomal Proteins, Non-Histone , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/isolation & purification , Imaging, Three-Dimensional , Mammals , Metaphase/genetics , Prophase/genetics
8.
Mol Cell ; 79(6): 917-933.e9, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32755595

ABSTRACT

Despite key roles in sister chromatid cohesion and chromosome organization, the mechanism by which cohesin rings are loaded onto DNA is still unknown. Here we combine biochemical approaches and cryoelectron microscopy (cryo-EM) to visualize a cohesin loading intermediate in which DNA is locked between two gates that lead into the cohesin ring. Building on this structural framework, we design experiments to establish the order of events during cohesin loading. In an initial step, DNA traverses an N-terminal kleisin gate that is first opened upon ATP binding and then closed as the cohesin loader locks the DNA against the ATPase gate. ATP hydrolysis will lead to ATPase gate opening to complete DNA entry. Whether DNA loading is successful or results in loop extrusion might be dictated by a conserved kleisin N-terminal tail that guides the DNA through the kleisin gate. Our results establish the molecular basis for cohesin loading onto DNA.


Subject(s)
Cell Cycle Proteins/ultrastructure , Chromatids/ultrastructure , Chromosomal Proteins, Non-Histone/ultrastructure , DNA/ultrastructure , Sister Chromatid Exchange/genetics , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation/genetics , Cryoelectron Microscopy , DNA/genetics , Nucleic Acid Conformation , Protein Conformation , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Cohesins
9.
Mol Cell ; 77(6): 1279-1293.e4, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32032532

ABSTRACT

Cohesin, a member of the SMC complex family, holds sister chromatids together but also shapes chromosomes by promoting the formation of long-range intra-chromatid loops, a process proposed to be mediated by DNA loop extrusion. Here we describe the roles of three cohesin partners, Pds5, Wpl1, and Eco1, in loop formation along either unreplicated or mitotic Saccharomyces cerevisiae chromosomes. Pds5 limits the size of DNA loops via two different pathways: the canonical Wpl1-mediated releasing activity and an Eco1-dependent mechanism. In the absence of Pds5, the main barrier to DNA loop expansion appears to be the centromere. Our data also show that Eco1 acetyl-transferase inhibits the translocase activity that powers loop formation and contributes to the positioning of loops through a mechanism that is distinguishable from its role in cohesion establishment. This study reveals that the mechanisms regulating cohesin-dependent chromatin loops are conserved among eukaryotes while promoting different functions.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Fungal/chemistry , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Acetyltransferases/genetics , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , Chromosomes, Fungal/genetics , Chromosomes, Fungal/metabolism , Mitosis , Nuclear Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Cohesins
10.
EMBO J ; 42(16): e113475, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37357575

ABSTRACT

Genetic information is stored in linear DNA molecules, which are highly folded inside cells. DNA replication along the folded template path yields two sister chromatids that initially occupy the same nuclear region in an intertwined arrangement. Dividing cells must disentangle and condense the sister chromatids into separate bodies such that a microtubule-based spindle can move them to opposite poles. While the spindle-mediated transport of sister chromatids has been studied in detail, the chromosome-intrinsic mechanics presegregating sister chromatids have remained elusive. Here, we show that human sister chromatids resolve extensively already during interphase, in a process dependent on the loop-extruding activity of cohesin, but not that of condensins. Increasing cohesin's looping capability increases sister DNA resolution in interphase nuclei to an extent normally seen only during mitosis, despite the presence of abundant arm cohesion. That cohesin can resolve sister chromatids so extensively in the absence of mitosis-specific activities indicates that DNA loop extrusion is a generic mechanism for segregating replicated genomes, shared across different Structural Maintenance of Chromosomes (SMC) protein complexes in all kingdoms of life.


Subject(s)
Chromatids , Chromosomal Proteins, Non-Histone , Humans , Chromatids/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Mitosis , DNA , G2 Phase , Cohesins
11.
Annu Rev Genet ; 53: 445-482, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31577909

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes are key organizers of chromosome architecture in all kingdoms of life. Despite seemingly divergent functions, such as chromosome segregation, chromosome maintenance, sister chromatid cohesion, and mitotic chromosome compaction, it appears that these complexes function via highly conserved mechanisms and that they represent a novel class of DNA translocases.


Subject(s)
Chromatids , Chromosomes/metabolism , DNA/chemistry , DNA/metabolism , Multiprotein Complexes/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cell Cycle Proteins/metabolism , Chromatids/chemistry , Chromatids/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Chromosomes/chemistry , Chromosomes/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Mitosis , Multiprotein Complexes/chemistry , Promoter Regions, Genetic , V(D)J Recombination , Cohesins
12.
Mol Cell ; 75(2): 224-237.e5, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31201089

ABSTRACT

Cohesin entraps sister DNAs within tripartite rings created by pairwise interactions between Smc1, Smc3, and Scc1. Because Smc1/3 ATPase heads can also interact with each other, cohesin rings have the potential to form a variety of sub-compartments. Using in vivo cysteine cross-linking, we show that when Smc1 and Smc3 ATPases are engaged in the presence of ATP (E heads), cohesin rings generate a "SMC (S) compartment" between hinge and E heads and a "kleisin (K) compartment" between E heads and their associated kleisin subunit. Upon ATP hydrolysis, cohesin's heads associate in a different mode, in which their signature motifs and their coiled coils are closely juxtaposed (J heads), creating alternative S and K compartments. We show that K compartments of either E or J type can entrap single DNAs, that acetylation of Smc3 during S phase is associated with J heads, and that sister DNAs are entrapped in J-K compartments.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , DNA/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Chromatids/genetics , DNA/chemistry , Dimerization , Models, Molecular , Protein Structure, Tertiary , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sister Chromatid Exchange/genetics , Cohesins
13.
Nat Rev Mol Cell Biol ; 15(9): 601-14, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25145851

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes, which in eukaryotic cells include cohesin, condensin and the Smc5/6 complex, are central regulators of chromosome dynamics and control sister chromatid cohesion, chromosome condensation, DNA replication, DNA repair and transcription. Even though the molecular mechanisms that lead to this large range of functions are still unclear, it has been established that the complexes execute their functions through their association with chromosomal DNA. A large set of data also indicates that SMC complexes work as intermolecular and intramolecular linkers of DNA. When combining these insights with results from ongoing analyses of their chromosomal binding, and how this interaction influences the structure and dynamics of chromosomes, a picture of how SMC complexes carry out their many functions starts to emerge.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomes, Human/metabolism , Multiprotein Complexes/metabolism , Animals , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone , Chromosomes, Human/genetics , DNA Repair/physiology , DNA Replication/physiology , Humans , Multiprotein Complexes/genetics , Transcription, Genetic/physiology
14.
Nature ; 586(7827): 139-144, 2020 10.
Article in English | MEDLINE | ID: mdl-32968280

ABSTRACT

The three-dimensional organization of the genome supports regulated gene expression, recombination, DNA repair, and chromosome segregation during mitosis. Chromosome conformation capture (Hi-C)1,2 analysis has revealed a complex genomic landscape of internal chromosomal structures in vertebrate cells3-7, but the identical sequence of sister chromatids has made it difficult to determine how they topologically interact in replicated chromosomes. Here we describe sister-chromatid-sensitive Hi-C (scsHi-C), which is based on labelling of nascent DNA with 4-thio-thymidine and nucleoside conversion chemistry. Genome-wide conformation maps of human chromosomes reveal that sister-chromatid pairs interact most frequently at the boundaries of topologically associating domains (TADs). Continuous loading of a dynamic cohesin pool separates sister-chromatid pairs inside TADs and is required to focus sister-chromatid contacts at TAD boundaries. We identified a subset of TADs that are overall highly paired and are characterized by facultative heterochromatin and insulated topological domains that form separately within individual sister chromatids. The rich pattern of sister-chromatid topologies and our scsHi-C technology will make it possible to investigate how physical interactions between identical DNA molecules contribute to DNA repair, gene expression, chromosome segregation, and potentially other biological processes.


Subject(s)
Chromatids/chemistry , Chromosome Pairing , DNA Replication , Genome, Human/genetics , Nucleic Acid Conformation , Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA/analysis , DNA/biosynthesis , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , Cohesins
15.
Mol Cell ; 70(6): 1134-1148.e7, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932904

ABSTRACT

Cohesin organizes DNA into chromatids, regulates enhancer-promoter interactions, and confers sister chromatid cohesion. Its association with chromosomes is regulated by hook-shaped HEAT repeat proteins that bind Scc1, namely Scc3, Pds5, and Scc2. Unlike Pds5, Scc2 is not a stable cohesin constituent but, as shown here, transiently replaces Pds5. Scc1 mutations that compromise its interaction with Scc2 adversely affect cohesin's ATPase activity and loading. Moreover, Scc2 mutations that alter how the ATPase responds to DNA abolish loading despite cohesin's initial association with loading sites. Lastly, Scc2 mutations that permit loading in the absence of Scc4 increase Scc2's association with chromosomal cohesin and reduce that of Pds5. We suggest that cohesin switches between two states: one with Pds5 bound that is unable to hydrolyze ATP efficiently but is capable of release from chromosomes and another in which Scc2 replaces Pds5 and stimulates ATP hydrolysis necessary for loading and translocation from loading sites.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , DNA, Fungal/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Cohesins
16.
Nucleic Acids Res ; 52(10): 5774-5791, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38597669

ABSTRACT

RAD51 filament is crucial for the homology-dependent repair of DNA double-strand breaks and stalled DNA replication fork protection. Positive and negative regulators control RAD51 filament assembly and disassembly. RAD51 is vital for genome integrity but excessive accumulation of RAD51 on chromatin causes genome instability and growth defects. However, the detailed mechanism underlying RAD51 disassembly by negative regulators and the physiological consequence of abnormal RAD51 persistence remain largely unknown. Here, we report the role of the human AAA+ ATPase FIGNL1 in suppressing a novel type of RAD51-mediated genome instability. FIGNL1 knockout human cells were defective in RAD51 dissociation after replication fork restart and accumulated ultra-fine chromosome bridges (UFBs), whose formation depends on RAD51 rather than replication fork stalling. FIGNL1 suppresses homologous recombination intermediate-like UFBs generated between sister chromatids at genomic loci with repeated sequences such as telomeres and centromeres. These data suggest that RAD51 persistence per se induces the formation of unresolved linkage between sister chromatids resulting in catastrophic genome instability. FIGNL1 facilitates post-replicative disassembly of RAD51 filament to suppress abnormal recombination intermediates and UFBs. These findings implicate FIGNL1 as a key factor required for active RAD51 removal after processing of stalled replication forks, which is essential to maintain genome stability.


Subject(s)
Genomic Instability , Rad51 Recombinase , Humans , Chromatids/metabolism , Chromatids/genetics , DNA Replication/genetics , Genomic Instability/genetics , Homologous Recombination/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Cell Line , Gene Knockout Techniques
17.
EMBO J ; 40(7): e106797, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33644892

ABSTRACT

Partitioning of the genome in meiosis occurs through two highly specialized cell divisions, named meiosis I and meiosis II. Step-wise cohesin removal is required for chromosome segregation in meiosis I, and sister chromatid segregation in meiosis II. In meiosis I, mono-oriented sister kinetochores appear as fused together when examined by high-resolution confocal microscopy, whereas they are clearly separated in meiosis II, when attachments are bipolar. It has been proposed that bipolar tension applied by the spindle is responsible for the physical separation of sister kinetochores, removal of cohesin protection, and chromatid separation in meiosis II. We show here that this is not the case, and initial separation of sister kinetochores occurs already in anaphase I independently of bipolar spindle forces applied on sister kinetochores, in mouse oocytes. This kinetochore individualization depends on separase cleavage activity. Crucially, without kinetochore individualization in meiosis I, bivalents when present in meiosis II oocytes separate into chromosomes and not sister chromatids. This shows that whether centromeric cohesin is removed or not is determined by the kinetochore structure prior to meiosis II.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Meiosis , Animals , Cells, Cultured , Chromatids/genetics , Chromatids/metabolism , Female , Mice , Mice, Inbred C57BL , Oocytes/metabolism , Cohesins
18.
J Cell Sci ; 136(11)2023 06 01.
Article in English | MEDLINE | ID: mdl-37288770

ABSTRACT

Sister chromatid cohesion is a multi-step process implemented throughout the cell cycle to ensure the correct transmission of chromosomes to daughter cells. Although cohesion establishment and mitotic cohesion dissolution have been extensively explored, the regulation of cohesin loading is still poorly understood. Here, we report that the methyltransferase NSD3 is essential for mitotic sister chromatid cohesion before mitosis entry. NSD3 interacts with the cohesin loader complex kollerin (composed of NIPBL and MAU2) and promotes the chromatin recruitment of MAU2 and cohesin at mitotic exit. We also show that NSD3 associates with chromatin in early anaphase, prior to the recruitment of MAU2 and RAD21, and dissociates from chromatin when prophase begins. Among the two NSD3 isoforms present in somatic cells, the long isoform is responsible for regulating kollerin and cohesin chromatin-loading, and its methyltransferase activity is required for efficient sister chromatid cohesion. Based on these observations, we propose that NSD3-dependent methylation contributes to sister chromatid cohesion by ensuring proper kollerin recruitment and thus cohesin loading.


Subject(s)
Cell Cycle Proteins , Chromatids , Histone Methyltransferases , Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromatids/metabolism , Chromatin , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Histone Methyltransferases/metabolism , Cohesins
19.
Mol Cell ; 67(4): 579-593.e6, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28781233

ABSTRACT

Precise control of sister chromatid separation during mitosis is pivotal to maintaining genomic integrity. Yet, the regulatory mechanisms involved are not well understood. Remarkably, we discovered that linker histone H1 phosphorylated at S/T18 decorated the inter-chromatid axial DNA on mitotic chromosomes. Sister chromatid resolution during mitosis required the eviction of such H1S/T18ph by the chaperone SET, with this process being independent of and most likely downstream of arm-cohesin dissociation. SET also directed the disassembly of Shugoshins in a polo-like kinase 1-augmented manner, aiding centromere resolution. SET ablation compromised mitotic fidelity as evidenced by unresolved sister chromatids with marked accumulation of H1S/T18ph and centromeric Shugoshin. Thus, chaperone-assisted eviction of linker histones and Shugoshins is a fundamental step in mammalian mitotic progression. Our findings also elucidate the functional implications of the decades-old observation of mitotic linker histone phosphorylation, serving as a paradigm to explore the role of linker histones in bio-signaling processes.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatids/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Mitosis , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromosome Segregation , DNA-Binding Proteins , Fibroblasts/metabolism , HEK293 Cells , Histone Chaperones/genetics , Humans , Mice , Oncogene Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Retinal Pigment Epithelium/metabolism , Signal Transduction , Transcription Factors/genetics , Transfection , Polo-Like Kinase 1
20.
Mol Cell ; 68(3): 605-614.e4, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100057

ABSTRACT

Cohesins establish sister chromatid cohesion during S phase and are removed when cohesin Scc1 is cleaved by separase at anaphase onset. During this process, cohesin Smc3 undergoes a cycle of acetylation: Smc3 acetylation by Eco1 in S phase stabilizes cohesin association with chromosomes, and its deacetylation by Hos1 in anaphase allows re-use of Smc3 in the next cell cycle. Here we find that Smc3 deacetylation by Hos1 has a more immediate effect in the early anaphase of budding yeast. Hos1 depletion significantly delayed sister chromatid separation and segregation. Smc3 deacetylation facilitated removal of cohesins from chromosomes without changing Scc1 cleavage efficiency, promoting dissolution of cohesion. This action is probably due to disengagement of Smc1-Smc3 heads prompted by de-repression of their ATPase activity. We suggest Scc1 cleavage per se is insufficient for efficient dissolution of cohesion in early anaphase; subsequent Smc3 deacetylation, triggered by Scc1 cleavage, is also required.


Subject(s)
Anaphase , Cell Cycle Proteins/metabolism , Chromatids/enzymology , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Histone Deacetylases/metabolism , Histone Demethylases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Acetylation , Cell Cycle Proteins/genetics , Chromatids/genetics , Chromosomal Proteins, Non-Histone/genetics , Histone Deacetylases/genetics , Histone Demethylases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Separase/genetics , Separase/metabolism , Signal Transduction , Time Factors , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL