Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
EMBO J ; 40(18): e108004, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34313341

ABSTRACT

Kinetochores form the link between chromosomes and microtubules of the mitotic spindle. The heterodecameric Dam1 complex (Dam1c) is a major component of the Saccharomyces cerevisiae outer kinetochore, assembling into 3 MDa-sized microtubule-embracing rings, but how ring assembly is specifically initiated in vivo remains to be understood. Here, we describe a molecular pathway that provides local control of ring assembly during the establishment of sister kinetochore bi-orientation. We show that Dam1c and the general microtubule plus end-associated protein (+TIP) Bim1/EB1 form a stable complex depending on a conserved motif in the Duo1 subunit of Dam1c. EM analyses reveal that Bim1 crosslinks protrusion domains of adjacent Dam1c heterodecamers and promotes the formation of oligomers with defined curvature. Disruption of the Dam1c-Bim1 interaction impairs kinetochore localization of Dam1c in metaphase and delays mitosis. Phosphorylation promotes Dam1c-Bim1 binding by relieving an intramolecular inhibition of the Dam1 C-terminus. In addition, Bim1 recruits Bik1/CLIP-170 to Dam1c and induces formation of full rings even in the absence of microtubules. Our data help to explain how new kinetochore end-on attachments are formed during the process of attachment error correction.


Subject(s)
Kinetochores/metabolism , Microtubule Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Saccharomycetales/physiology , Chromosome Segregation , Mitosis/physiology , Multiprotein Complexes/metabolism , Phosphorylation , Protein Binding , Spindle Apparatus/metabolism
2.
J Cell Biol ; 216(11): 3785-3798, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28912124

ABSTRACT

Cell spreading requires the coupling of actin-driven membrane protrusion and integrin-mediated adhesion to the extracellular matrix. The integrin-activating adaptor protein kindlin-2 plays a central role for cell adhesion and membrane protrusion by directly binding and recruiting paxillin to nascent adhesions. Here, we report that kindlin-2 has a dual role during initial cell spreading: it binds paxillin via the pleckstrin homology and F0 domains to activate Rac1, and it directly associates with the Arp2/3 complex to induce Rac1-mediated membrane protrusions. Consistently, abrogation of kindlin-2 binding to Arp2/3 impairs lamellipodia formation and cell spreading. Our findings identify kindlin-2 as a key protein that couples cell adhesion by activating integrins and the induction of membrane protrusions by activating Rac1 and supplying Rac1 with the Arp2/3 complex.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Cell Adhesion , Cell Shape , Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , Muscle Proteins/metabolism , Paxillin/metabolism , Pseudopodia/metabolism , Actin-Related Protein 2-3 Complex/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Genotype , Mice, Knockout , Muscle Proteins/deficiency , Muscle Proteins/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Paxillin/genetics , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Talin/deficiency , Talin/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
3.
J Cell Biol ; 216(4): 961-981, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28320825

ABSTRACT

Kinetochores are macromolecular assemblies that connect chromosomes to spindle microtubules (MTs) during mitosis. The metazoan-specific ≈800-kD ROD-Zwilch-ZW10 (RZZ) complex builds a fibrous corona that assembles on mitotic kinetochores before MT attachment to promote chromosome alignment and robust spindle assembly checkpoint signaling. In this study, we combine biochemical reconstitutions, single-particle electron cryomicroscopy, cross-linking mass spectrometry, and structural modeling to build a complete model of human RZZ. We find that RZZ is structurally related to self-assembling cytosolic coat scaffolds that mediate membrane cargo trafficking, including Clathrin, Sec13-Sec31, and αß'ε-COP. We show that Spindly, a dynein adaptor, is related to BicD2 and binds RZZ directly in a farnesylation-dependent but membrane-independent manner. Through a targeted chemical biology approach, we identify ROD as the Spindly farnesyl receptor. Our results suggest that RZZ is dynein's cargo at human kinetochores.


Subject(s)
Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/physiology , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Dyneins/metabolism , HeLa Cells , Humans , Kinetochores/physiology , M Phase Cell Cycle Checkpoints/physiology , Microtubules/metabolism , Mitosis/physiology , Protein Transport/physiology
4.
Elife ; 52016 11 30.
Article in English | MEDLINE | ID: mdl-27901467

ABSTRACT

The Hsp104 disaggregase is a two-ring ATPase machine that rescues various forms of non-native proteins including the highly resistant amyloid fibers. The structural-mechanistic underpinnings of how the recovery of toxic protein aggregates is promoted and how this potent unfolding activity is prevented from doing collateral damage to cellular proteins are not well understood. Here, we present structural and biochemical data revealing the organization of Hsp104 from Chaetomium thermophilum at 3.7 Å resolution. We show that the coiled-coil domains encircling the disaggregase constitute a 'restraint mask' that sterically controls the mobility and thus the unfolding activity of the ATPase modules. In addition, we identify a mechanical linkage that coordinates the activity of the two ATPase rings and accounts for the high unfolding potential of Hsp104. Based on these findings, we propose a general model for how Hsp104 and related chaperones operate and are kept under control until recruited to appropriate substrates.


Subject(s)
Chaetomium/enzymology , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Aggregates , Protein Conformation , Protein Domains , Protein Unfolding
5.
Cell ; 167(4): 1028-1040.e15, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27881301

ABSTRACT

Kinetochores, multisubunit protein assemblies, connect chromosomes to spindle microtubules to promote chromosome segregation. The 10-subunit KMN assembly (comprising KNL1, MIS12, and NDC80 complexes, designated KNL1C, MIS12C, and NDC80C) binds microtubules and regulates mitotic checkpoint function through NDC80C and KNL1C, respectively. MIS12C, on the other hand, connects the KMN to the chromosome-proximal domain of the kinetochore through a direct interaction with CENP-C. The structural basis for this crucial bridging function of MIS12C is unknown. Here, we report crystal structures of human MIS12C associated with a fragment of CENP-C and unveil the role of Aurora B kinase in the regulation of this interaction. The structure of MIS12:CENP-C complements previously determined high-resolution structures of functional regions of NDC80C and KNL1C and allows us to build a near-complete structural model of the KMN assembly. Our work illuminates the structural organization of essential chromosome segregation machinery that is conserved in most eukaryotes.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Crystallography, X-Ray , Kinetochores/chemistry , Multiprotein Complexes/chemistry , Animals , Aurora Kinase B/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cytoskeletal Proteins , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Models, Chemical , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism
6.
Nucleus ; 7(5): 476-484, 2016 Sep 02.
Article in English | MEDLINE | ID: mdl-27676121

ABSTRACT

Chromatin structure and function are determined by a plethora of proteins whose genome-wide distribution is typically assessed by immunoprecipitation (ChIP). Here, we developed a novel tool to investigate the local chromatin environment at specific DNA sequences. We combined the programmable DNA binding of dCas9 with the promiscuous biotin ligase BirA* (CasID) to biotinylate proteins in the direct vicinity of specific loci. Subsequent streptavidin-mediated precipitation and mass spectrometry identified both known and previously unknown chromatin factors associated with repetitive telomeric, major satellite and minor satellite DNA. With super-resolution microscopy, we confirmed the localization of the putative transcription factor ZNF512 at chromocenters. The versatility of CasID facilitates the systematic elucidation of functional protein complexes and locus-specific chromatin composition.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Chromatin/chemistry , Chromatin/metabolism , Animals , Base Sequence , Cell Line , Chromatin/genetics , DNA/genetics , DNA/metabolism , Genomics , Mass Spectrometry , Mice , Microscopy, Fluorescence
7.
Nat Commun ; 7: 12523, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27549742

ABSTRACT

The cohesin subunits Smc1, Smc3 and Scc1 form large tripartite rings which mediate sister chromatid cohesion and chromatin structure. These are thought to entrap DNA with the help of the associated proteins SA1/2 and Pds5A/B. Structural information is available for parts of cohesin, but analyses of entire cohesin complexes are limited by their flexibility. Here we generated a more rigid 'bonsai' cohesin by truncating the coiled coils of Smc1 and Smc3 and used single-particle electron microscopy, chemical crosslinking-mass spectrometry and in silico modelling to generate three-dimensional models of cohesin bound to Pds5B. The HEAT-repeat protein Pds5B forms a curved structure around the nucleotide-binding domains of Smc1 and Smc3 and bridges the Smc3-Scc1 and SA1-Scc1 interfaces. These results indicate that Pds5B forms an integral part of the cohesin ring by contacting all other cohesin subunits, a property that may reflect the complex role of Pds5 proteins in controlling cohesin-DNA interactions.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Protein Engineering , Transcription Factors/metabolism , Cell Cycle Proteins/ultrastructure , Chromosomal Proteins, Non-Histone/ultrastructure , Computer Simulation , Humans , Models, Molecular , Protein Binding , Protein Domains , Cohesins
8.
Elife ; 52016 06 10.
Article in English | MEDLINE | ID: mdl-27282391

ABSTRACT

Transcription regulation in metazoans often involves promoter-proximal pausing of RNA polymerase (Pol) II, which requires the 4-subunit negative elongation factor (NELF). Here we discern the functional architecture of human NELF through X-ray crystallography, protein crosslinking, biochemical assays, and RNA crosslinking in cells. We identify a NELF core subcomplex formed by conserved regions in subunits NELF-A and NELF-C, and resolve its crystal structure. The NELF-AC subcomplex binds single-stranded nucleic acids in vitro, and NELF-C associates with RNA in vivo. A positively charged face of NELF-AC is involved in RNA binding, whereas the opposite face of the NELF-AC subcomplex binds NELF-B. NELF-B is predicted to form a HEAT repeat fold, also binds RNA in vivo, and anchors the subunit NELF-E, which is confirmed to bind RNA in vivo. These results reveal the three-dimensional architecture and three RNA-binding faces of NELF.


Subject(s)
RNA/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Crystallography, X-Ray , Gene Expression Regulation , Humans , Models, Molecular , Protein Binding , Protein Conformation , Transcription, Genetic
9.
Open Biol ; 6(2): 150236, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26911624

ABSTRACT

Accurate chromosome segregation during mitosis and meiosis is crucial for cellular and organismal viability. Kinetochores connect chromosomes with spindle microtubules and are essential for chromosome segregation. These large protein scaffolds emerge from the centromere, a specialized region of the chromosome enriched with the histone H3 variant CENP-A. In most eukaryotes, the kinetochore core consists of the centromere-proximal constitutive centromere-associated network (CCAN), which binds CENP-A and contains 16 subunits, and of the centromere-distal Knl1 complex, Mis12 complex, Ndc80 complex (KMN) network, which binds microtubules and contains 10 subunits. In the fruitfly, Drosophila melanogaster, the kinetochore underwent remarkable simplifications. All CCAN subunits, with the exception of centromeric protein C (CENP-C), and two KMN subunits, Dsn1 and Zwint, cannot be identified in this organism. In addition, two paralogues of the KMN subunit Nnf1 (Nnf1a and Nnf1b) are present. Finally, the Spc105R subunit, homologous to human Knl1/CASC5, underwent considerable sequence changes in comparison with other organisms. We combined biochemical reconstitution with biophysical and structural methods to investigate how these changes reflect on the organization of the Drosophila KMN network. We demonstrate that the Nnf1a and Nnf1b paralogues are subunits of distinct complexes, both of which interact directly with Spc105R and with CENP-C, for the latter of which we identify a binding site on the Mis12 subunit. Our studies shed light on the structural and functional organization of a highly divergent kinetochore particle.


Subject(s)
Drosophila melanogaster/metabolism , Kinetochores/metabolism , Amino Acid Sequence , Animals , Binding Sites , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Drosophila melanogaster/genetics , Humans , Kinetochores/chemistry , Kinetochores/ultrastructure , Mass Spectrometry , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins , Sequence Alignment
10.
Nucleic Acids Res ; 43(17): e112, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26007658

ABSTRACT

Any profound comprehension of gene function requires detailed information about the subcellular localization, molecular interactions and spatio-temporal dynamics of gene products. We developed a multifunctional integrase (MIN) tag for rapid and versatile genome engineering that serves not only as a genetic entry site for the Bxb1 integrase but also as a novel epitope tag for standardized detection and precipitation. For the systematic study of epigenetic factors, including Dnmt1, Dnmt3a, Dnmt3b, Tet1, Tet2, Tet3 and Uhrf1, we generated MIN-tagged embryonic stem cell lines and created a toolbox of prefabricated modules that can be integrated via Bxb1-mediated recombination. We used these functional modules to study protein interactions and their spatio-temporal dynamics as well as gene expression and specific mutations during cellular differentiation and in response to external stimuli. Our genome engineering strategy provides a versatile open platform for efficient generation of multiple isogenic cell lines to study gene function under physiological conditions.


Subject(s)
Cell Engineering/methods , Animals , Antibodies, Monoclonal , CRISPR-Cas Systems , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/metabolism , Gene Expression , Genetic Loci , Genomics/methods , Integrases/genetics , Integrases/immunology , Integrases/metabolism , Mutation , Rats , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...