Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cell ; 165(6): 1440-1453, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259151

ABSTRACT

Protein ubiquitination involves E1, E2, and E3 trienzyme cascades. E2 and RING E3 enzymes often collaborate to first prime a substrate with a single ubiquitin (UB) and then achieve different forms of polyubiquitination: multiubiquitination of several sites and elongation of linkage-specific UB chains. Here, cryo-EM and biochemistry show that the human E3 anaphase-promoting complex/cyclosome (APC/C) and its two partner E2s, UBE2C (aka UBCH10) and UBE2S, adopt specialized catalytic architectures for these two distinct forms of polyubiquitination. The APC/C RING constrains UBE2C proximal to a substrate and simultaneously binds a substrate-linked UB to drive processive multiubiquitination. Alternatively, during UB chain elongation, the RING does not bind UBE2S but rather lures an evolving substrate-linked UB to UBE2S positioned through a cullin interaction to generate a Lys11-linked chain. Our findings define mechanisms of APC/C regulation, and establish principles by which specialized E3-E2-substrate-UB architectures control different forms of polyubiquitination.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/chemistry , Anaphase-Promoting Complex-Cyclosome/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Biocatalysis , Cryoelectron Microscopy , Humans , Models, Molecular , Saccharomyces cerevisiae Proteins/chemistry , Structure-Activity Relationship , Ubiquitination
2.
Mol Cell ; 77(5): 1092-1106.e9, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31973889

ABSTRACT

Co-opting Cullin4 RING ubiquitin ligases (CRL4s) to inducibly degrade pathogenic proteins is emerging as a promising therapeutic strategy. Despite intense efforts to rationally design degrader molecules that co-opt CRL4s, much about the organization and regulation of these ligases remains elusive. Here, we establish protein interaction kinetics and estimation of stoichiometries (PIKES) analysis, a systematic proteomic profiling platform that integrates cellular engineering, affinity purification, chemical stabilization, and quantitative mass spectrometry to investigate the dynamics of interchangeable multiprotein complexes. Using PIKES, we show that ligase assemblies of Cullin4 with individual substrate receptors differ in abundance by up to 200-fold and that Cand1/2 act as substrate receptor exchange factors. Furthermore, degrader molecules can induce the assembly of their cognate CRL4, and higher expression of the associated substrate receptor enhances degrader potency. Beyond the CRL4 network, we show how PIKES can reveal systems level biochemistry for cellular protein networks important to drug development.


Subject(s)
Chromatography, High Pressure Liquid , Proteomics/methods , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Ubiquitin-Protein Ligases/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Protein Interaction Maps , Proteolysis , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
3.
Nat Chem Biol ; 20(1): 93-102, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37679459

ABSTRACT

Molecular glue degraders are an effective therapeutic modality, but their design principles are not well understood. Recently, several unexpectedly diverse compounds were reported to deplete cyclin K by linking CDK12-cyclin K to the DDB1-CUL4-RBX1 E3 ligase. Here, to investigate how chemically dissimilar small molecules trigger cyclin K degradation, we evaluated 91 candidate degraders in structural, biophysical and cellular studies and reveal all compounds acquire glue activity via simultaneous CDK12 binding and engagement of DDB1 interfacial residues, in particular Arg928. While we identify multiple published kinase inhibitors as cryptic degraders, we also show that these glues do not require pronounced inhibitory properties for activity and that the relative degree of CDK12 inhibition versus cyclin K degradation is tuneable. We further demonstrate cyclin K degraders have transcriptional signatures distinct from CDK12 inhibitors, thereby offering unique therapeutic opportunities. The systematic structure-activity relationship analysis presented herein provides a conceptual framework for rational molecular glue design.


Subject(s)
Cyclins , Ubiquitin-Protein Ligases , Cyclins/metabolism , Ubiquitin-Protein Ligases/metabolism , Proteolysis , Structure-Activity Relationship
4.
Nature ; 585(7824): 293-297, 2020 09.
Article in English | MEDLINE | ID: mdl-32494016

ABSTRACT

Molecular glue compounds induce protein-protein interactions that, in the context of a ubiquitin ligase, lead to protein degradation1. Unlike traditional enzyme inhibitors, these molecular glue degraders act substoichiometrically to catalyse the rapid depletion of previously inaccessible targets2. They are clinically effective and highly sought-after, but have thus far only been discovered serendipitously. Here, through systematically mining databases for correlations between the cytotoxicity of 4,518 clinical and preclinical small molecules and the expression levels of E3 ligase components across hundreds of human cancer cell lines3-5, we identify CR8-a cyclin-dependent kinase (CDK) inhibitor6-as a compound that acts as a molecular glue degrader. The CDK-bound form of CR8 has a solvent-exposed pyridyl moiety that induces the formation of a complex between CDK12-cyclin K and the CUL4 adaptor protein DDB1, bypassing the requirement for a substrate receptor and presenting cyclin K for ubiquitination and degradation. Our studies demonstrate that chemical alteration of surface-exposed moieties can confer gain-of-function glue properties to an inhibitor, and we propose this as a broader strategy through which target-binding molecules could be converted into molecular glues.


Subject(s)
Cyclins/deficiency , Cyclins/metabolism , Proteolysis/drug effects , Purines/chemistry , Purines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Cyclins/chemistry , DNA-Binding Proteins/metabolism , Humans , Models, Molecular , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Purines/toxicity , Pyridines/toxicity , Small Molecule Libraries/analysis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Ubiquitination/drug effects
5.
Nat Chem Biol ; 16(11): 1199-1207, 2020 11.
Article in English | MEDLINE | ID: mdl-32747809

ABSTRACT

Targeted protein degradation is a new therapeutic modality based on drugs that destabilize proteins by inducing their proximity to E3 ubiquitin ligases. Of particular interest are molecular glues that can degrade otherwise unligandable proteins by orchestrating direct interactions between target and ligase. However, their discovery has so far been serendipitous, thus hampering broad translational efforts. Here, we describe a scalable strategy toward glue degrader discovery that is based on chemical screening in hyponeddylated cells coupled to a multi-omics target deconvolution campaign. This approach led us to identify compounds that induce ubiquitination and degradation of cyclin K by prompting an interaction of CDK12-cyclin K with a CRL4B ligase complex. Notably, this interaction is independent of a dedicated substrate receptor, thus functionally segregating this mechanism from all described degraders. Collectively, our data outline a versatile and broadly applicable strategy to identify degraders with nonobvious mechanisms and thus empower future drug discovery efforts.


Subject(s)
Acetamides/chemistry , Anti-Bacterial Agents/pharmacology , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Doxycycline/pharmacology , Hydrazines/chemistry , Indoles/chemistry , Proteolysis/drug effects , Retinoblastoma-Binding Protein 7/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gene Expression Regulation , Humans , Molecular Structure , Protein Binding , Protein Conformation , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
6.
Nature ; 532(7597): 127-30, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26909574

ABSTRACT

Thalidomide and its derivatives, lenalidomide and pomalidomide, are immune modulatory drugs (IMiDs) used in the treatment of haematologic malignancies. IMiDs bind CRBN, the substrate receptor of the CUL4-RBX1-DDB1-CRBN (also known as CRL4(CRBN)) E3 ubiquitin ligase, and inhibit ubiquitination of endogenous CRL4(CRBN) substrates. Unexpectedly, IMiDs also repurpose the ligase to target new proteins for degradation. Lenalidomide induces degradation of the lymphoid transcription factors Ikaros and Aiolos (also known as IKZF1 and IKZF3), and casein kinase 1α (CK1α), which contributes to its clinical efficacy in the treatment of multiple myeloma and 5q-deletion associated myelodysplastic syndrome (del(5q) MDS), respectively. How lenalidomide alters the specificity of the ligase to degrade these proteins remains elusive. Here we present the 2.45 Å crystal structure of DDB1-CRBN bound to lenalidomide and CK1α. CRBN and lenalidomide jointly provide the binding interface for a CK1α ß-hairpin-loop located in the kinase N-lobe. We show that CK1α binding to CRL4(CRBN) is strictly dependent on the presence of an IMiD. Binding of IKZF1 to CRBN similarly requires the compound and both, IKZF1 and CK1α, use a related binding mode. Our study provides a mechanistic explanation for the selective efficacy of lenalidomide in del(5q) MDS therapy. We anticipate that high-affinity protein-protein interactions induced by small molecules will provide opportunities for drug development, particularly for targeted protein degradation.


Subject(s)
Casein Kinase Ialpha/metabolism , Thalidomide/analogs & derivatives , Ubiquitin-Protein Ligases/metabolism , Binding Sites/drug effects , Casein Kinase Ialpha/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Ikaros Transcription Factor/chemistry , Ikaros Transcription Factor/metabolism , Lenalidomide , Models, Molecular , Protein Binding/drug effects , Proteolysis/drug effects , Structure-Activity Relationship , Substrate Specificity/drug effects , Thalidomide/chemistry , Thalidomide/metabolism , Thalidomide/pharmacology , Ubiquitin-Protein Ligases/chemistry , Ubiquitination/drug effects
7.
Nature ; 531(7596): 598-603, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029275

ABSTRACT

The cullin-RING ubiquitin E3 ligase (CRL) family comprises over 200 members in humans. The COP9 signalosome complex (CSN) regulates CRLs by removing their ubiquitin-like activator NEDD8. The CUL4A-RBX1-DDB1-DDB2 complex (CRL4A(DDB2)) monitors the genome for ultraviolet-light-induced DNA damage. CRL4A(DBB2) is inactive in the absence of damaged DNA and requires CSN to regulate the repair process. The structural basis of CSN binding to CRL4A(DDB2) and the principles of CSN activation are poorly understood. Here we present cryo-electron microscopy structures for CSN in complex with neddylated CRL4A ligases to 6.4 Å resolution. The CSN conformers defined by cryo-electron microscopy and a novel apo-CSN crystal structure indicate an induced-fit mechanism that drives CSN activation by neddylated CRLs. We find that CSN and a substrate cannot bind simultaneously to CRL4A, favouring a deneddylated, inactive state for substrate-free CRL4 complexes. These architectural and regulatory principles appear conserved across CRL families, allowing global regulation by CSN.


Subject(s)
Biocatalysis , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Peptide Hydrolases/metabolism , Peptide Hydrolases/ultrastructure , Allosteric Regulation , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Binding Sites , COP9 Signalosome Complex , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Carrier Proteins/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , Cullin Proteins/chemistry , Cullin Proteins/metabolism , Cullin Proteins/ultrastructure , DNA Damage , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Humans , Kinetics , Models, Molecular , Multiprotein Complexes/chemistry , Peptide Hydrolases/chemistry , Protein Binding , Ubiquitination , Ubiquitins/metabolism
8.
Mol Cell ; 56(2): 246-260, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25306923

ABSTRACT

Polyubiquitination by E2 and E3 enzymes is a predominant mechanism regulating protein function. Some RING E3s, including anaphase-promoting complex/cyclosome (APC), catalyze polyubiquitination by sequential reactions with two different E2s. An initiating E2 ligates ubiquitin to an E3-bound substrate. Another E2 grows a polyubiquitin chain on the ubiquitin-primed substrate through poorly defined mechanisms. Here we show that human APC's RING domain is repurposed for dual functions in polyubiquitination. The canonical RING surface activates an initiating E2-ubiquitin intermediate for substrate modification. However, APC engages and activates its specialized ubiquitin chain-elongating E2 UBE2S in ways that differ from current paradigms. During chain assembly, a distinct APC11 RING surface helps deliver a substrate-linked ubiquitin to accept another ubiquitin from UBE2S. Our data define mechanisms of APC/UBE2S-mediated polyubiquitination, reveal diverse functions of RING E3s and E2s, and provide a framework for understanding distinctive RING E3 features specifying ubiquitin chain elongation.


Subject(s)
Apc11 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apc2 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Peptide Biosynthesis, Nucleic Acid-Independent , Polyubiquitin/biosynthesis , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination/physiology , Amino Acid Sequence , Apc4 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle Checkpoints , HeLa Cells , Humans , Molecular Sequence Data , Polyubiquitin/genetics , Protein Structure, Tertiary
9.
Proc Natl Acad Sci U S A ; 113(19): E2564-9, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27114506

ABSTRACT

Analyses of protein complexes are facilitated by methods that enable the generation of recombinant complexes via coexpression of their subunits from multigene DNA constructs. However, low experimental throughput limits the generation of such constructs in parallel. Here we describe a method that allows up to 25 cDNAs to be assembled into a single baculoviral expression vector in only two steps. This method, called biGBac, uses computationally optimized DNA linker sequences that enable the efficient assembly of linear DNA fragments, using reactions developed by Gibson for the generation of synthetic genomes. The biGBac method uses a flexible and modular "mix and match" approach and enables the generation of baculoviruses from DNA constructs at any assembly stage. Importantly, it is simple, efficient, and fast enough to allow the manual generation of many multigene expression constructs in parallel. We have used this method to generate and characterize recombinant forms of the anaphase-promoting complex/cyclosome, cohesin, and kinetochore complexes.


Subject(s)
Baculoviridae/genetics , Genetic Vectors/genetics , Multigene Family/genetics , Multiprotein Complexes/genetics , Protein Engineering/methods , Transfection/methods , Cloning, Molecular/methods , Multiprotein Complexes/metabolism , Mutagenesis, Site-Directed/methods , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
10.
EMBO J ; 33(22): 2643-58, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25257309

ABSTRACT

Although splicing is essential for the expression of most eukaryotic genes, inactivation of splicing factors causes specific defects in mitosis. The molecular cause of this defect is unknown. Here, we show that the spliceosome subunits SNW1 and PRPF8 are essential for sister chromatid cohesion in human cells. A transcriptome-wide analysis revealed that SNW1 or PRPF8 depletion affects the splicing of specific introns in a subset of pre-mRNAs, including pre-mRNAs encoding the cohesion protein sororin and the APC/C subunit APC2. SNW1 depletion causes cohesion defects predominantly by reducing sororin levels, which causes destabilisation of cohesin on DNA. SNW1 depletion also reduces APC/C activity and contributes to cohesion defects indirectly by delaying mitosis and causing "cohesion fatigue". Simultaneous expression of sororin and APC2 from intron-less cDNAs restores cohesion in SNW1-depleted cells. These results indicate that the spliceosome is required for mitosis because it enables expression of genes essential for cohesion. Our transcriptome-wide identification of retained introns in SNW1- and PRPF8-depleted cells may help to understand the aetiology of diseases associated with splicing defects, such as retinosa pigmentosum and cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Chromatids/metabolism , Cytoskeletal Proteins/metabolism , Nuclear Receptor Coactivators/metabolism , RNA Precursors/metabolism , RNA Splicing/physiology , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Chromatids/genetics , Cytoskeletal Proteins/genetics , Gene Deletion , HeLa Cells , Humans , Nuclear Receptor Coactivators/genetics , RNA Precursors/genetics , Transcriptome/physiology
11.
Nat Methods ; 12(9): 859-65, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26237227

ABSTRACT

Molecular machines or macromolecular complexes are supramolecular assemblies of biomolecules with a variety of functions. Structure determination of these complexes in a purified state is often tedious owing to their compositional complexity and the associated relative structural instability. To improve the stability of macromolecular complexes in vitro, we present a generic method that optimizes the stability, homogeneity and solubility of macromolecular complexes by sparse-matrix screening of their thermal unfolding behavior in the presence of various buffers and small molecules. The method includes the automated analysis of thermal unfolding curves based on a biophysical unfolding model for complexes. We found that under stabilizing conditions, even large multicomponent complexes reveal an almost ideal two-state unfolding behavior. We envisage an improved biochemical understanding of purified macromolecules as well as a substantial boost in successful macromolecular complex structure determination by both X-ray crystallography and cryo-electron microscopy.


Subject(s)
Algorithms , Models, Chemical , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Software , Binding Sites , Computer Simulation , Crystallization , Protein Binding , Protein Conformation , Protein Folding
13.
Nat Chem Biol ; 13(5): 452-453, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28406888

Subject(s)
Proteolysis
14.
ACS Chem Biol ; 17(1): 24-31, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34982531

ABSTRACT

We successfully repurpose the DNA repair protein methylguanine methyltransferase (MGMT) as an inducible degron for protein fusions. MGMT is a suicide protein that removes alkyl groups from the O6 position of guanine (O6G) and is thereafter quickly degraded by the ubiquitin proteasome pathway (UPP). Starting with MGMT pseudosubstrates (benzylguanine and lomeguatrib), we first demonstrate that these lead to potent MGMT depletion while affecting little else in the proteome. We then show that fusion proteins of MGMT undergo rapid UPP-dependent degradation in response to pseudosubstrates. Mechanistic studies confirm the involvement of the UPP, while revealing that at least two E3 ligase classes can degrade MGMT depending on cell-line and expression type (native or ectopic). We also demonstrate the technique's versatility with two clinically relevant examples: degradation of KRASG12C and a chimeric antigen receptor.


Subject(s)
DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , Tumor Suppressor Proteins/metabolism , CRISPR-Cas Systems , Cell Line , DNA Damage , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/genetics , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/genetics , Humans , Ligands , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
15.
Cell Chem Biol ; 27(1): 16-18, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31951816

ABSTRACT

PCSK9 heightens LDL cholesterol levels by chaperoning the liver LDL receptor to lysosomes for degradation. In this issue of Cell Chemical Biology, Petrilli et al. (2020) identify novel PCSK9 ligands and convert them into a proof-of-concept degrader, offering a unique way to modulate this key protein-protein interaction.


Subject(s)
Proprotein Convertase 9 , Cholesterol, LDL , Ligands , Proteolysis
16.
Science ; 362(6414)2018 11 02.
Article in English | MEDLINE | ID: mdl-30385546

ABSTRACT

The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. We screened the human C2H2 zinc finger proteome for degradation in the presence of thalidomide analogs, identifying 11 zinc finger degrons. Structural and functional characterization of the C2H2 zinc finger degrons demonstrates how diverse zinc finger domains bind the permissive drug-CRBN interface. Computational zinc finger docking and biochemical analysis predict that more than 150 zinc fingers bind the drug-CRBN complex in vitro, and we show that selective zinc finger degradation can be achieved through compound modifications. Our results provide a rationale for therapeutically targeting transcription factors that were previously considered undruggable.


Subject(s)
CYS2-HIS2 Zinc Fingers , Lenalidomide/pharmacology , Peptide Hydrolases/metabolism , Proteolysis/drug effects , Thalidomide/analogs & derivatives , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , HEK293 Cells , Humans , Ikaros Transcription Factor/metabolism , Proteome/metabolism , Thalidomide/pharmacology
17.
Article in English | MEDLINE | ID: mdl-17565194

ABSTRACT

The gram-positive bacterium Bacillus subtilis contains three proteins belonging to the signal recognition particle (SRP) type GTPase family. The well characterized signal sequence-binding protein SRP54 and the SRP receptor protein FtsY are universally conserved components of the SRP system of protein transport. The third member, FlhF, has been implicated in the placement and assembly of polar flagella. This article describes the overexpression and preliminary X-ray crystallographic analysis of an FlhF fragment that corresponds to the well characterized GTPase domains in SRP54 and FtsY. Three crystal forms are reported with either GDP or GMPPNP and diffract to a resolution of about 3 A.


Subject(s)
Bacillus subtilis/chemistry , Bacterial Proteins/chemistry , Monomeric GTP-Binding Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/isolation & purification , Protein Conformation
18.
Curr Biol ; 24(19): 2228-37, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25220052

ABSTRACT

BACKGROUND: Cohesin mediates sister chromatid cohesion by topologically entrapping sister DNA molecules inside its ring structure. Cohesin is loaded onto DNA by the Scc2/NIPBL-Scc4/MAU2-loading complex in a manner that depends on the adenosine triphosphatase (ATPase) activity of cohesin's Smc1 and Smc3 subunits. Subsequent cohesion establishment during DNA replication depends on Smc3 acetylation by Esco1 and Esco2 and on recruitment of sororin, which "locks" cohesin on DNA by inactivating the cohesin release factor Wapl. RESULTS: Human cohesin ATPase mutants associate transiently with DNA in a manner that depends on the loading complex but cannot be stabilized on chromatin by depletion of Wapl. These mutants cannot be acetylated, fail to interact with sororin, and do not mediate cohesion. The absence of Smc3 acetylation in the ATPase mutants is not a consequence of their transient association with DNA but is directly caused by their inability to hydrolyze ATP because acetylation of wild-type cohesin also depends on ATP hydrolysis. CONCLUSIONS: Our data indicate that cohesion establishment involves the following steps. First, cohesin transiently associates with DNA in a manner that depends on the loading complex. Subsequently, ATP hydrolysis by cohesin leads to entrapment of DNA and converts Smc3 into a state that can be acetylated. Finally, Smc3 acetylation leads to recruitment of sororin, inhibition of Wapl, and stabilization of cohesin on DNA. Our finding that cohesin's ATPase activity is required for both cohesin loading and Smc3 acetylation raises the possibility that cohesion establishment is directly coupled to the reaction in which cohesin entraps DNA.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Humans , Hydrolysis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Proteins/genetics , Proteins/metabolism , Sister Chromatid Exchange , Cohesins
19.
Nat Struct Mol Biol ; 20(7): 827-35, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23708605

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) is a ~1.5-MDa multiprotein E3 ligase enzyme that regulates cell division by promoting timely ubiquitin-mediated proteolysis of key cell-cycle regulatory proteins. Inhibition of human APC/C(CDH1) during interphase by early mitotic inhibitor 1 (EMI1) is essential for accurate coordination of DNA synthesis and mitosis. Here, we report a hybrid structural approach involving NMR, electron microscopy and enzymology, which reveal that EMI1's 143-residue C-terminal domain inhibits multiple APC/C(CDH1) functions. The intrinsically disordered D-box, linker and tail elements, together with a structured zinc-binding domain, bind distinct regions of APC/C(CDH1) to synergistically both block the substrate-binding site and inhibit ubiquitin-chain elongation. The functional importance of intrinsic structural disorder is explained by enabling a small inhibitory domain to bind multiple sites to shut down various functions of a 'molecular machine' nearly 100 times its size.


Subject(s)
Cadherins/chemistry , Cell Cycle Proteins/chemistry , F-Box Proteins/chemistry , Ubiquitin-Protein Ligase Complexes/chemistry , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Antigens, CD , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/pharmacology , Cell Cycle Proteins/ultrastructure , F-Box Proteins/metabolism , F-Box Proteins/pharmacology , F-Box Proteins/ultrastructure , Humans , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Folding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship , Substrate Specificity , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligase Complexes/ultrastructure , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitinated Proteins/metabolism , Ubiquitination , Ultracentrifugation
20.
Nat Struct Mol Biol ; 19(11): 1116-23, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23007861

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) bound to CDC20 (APC/C(CDC20)) initiates anaphase by ubiquitylating B-type cyclins and securin. During chromosome bi-orientation, CDC20 assembles with MAD2, BUBR1 and BUB3 into a mitotic checkpoint complex (MCC) that inhibits substrate recruitment to the APC/C. APC/C activation depends on MCC disassembly, which was proposed to require CDC20 autoubiquitylation. Here we characterize APC15, a human APC/C subunit related to yeast Mnd2. APC15 is located near APC/C's MCC binding site; it is required for APC/C-bound MCC (APC/C(MCC))-dependent CDC20 autoubiquitylation and degradation and for timely anaphase initiation but is dispensable for substrate ubiquitylation by APC/C(CDC20) and APC/C(CDH1). Our results support the model wherein MCC is continuously assembled and disassembled to enable rapid activation of APC/C(CDC20) and CDC20 autoubiquitylation promotes MCC disassembly. We propose that APC15 and Mnd2 negatively regulate APC/C coactivators and report generation of recombinant human APC/C.


Subject(s)
Cell Cycle Proteins/metabolism , M Phase Cell Cycle Checkpoints/physiology , Models, Biological , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Calcium-Binding Proteins/metabolism , Cdc20 Proteins , Cell Cycle Proteins/genetics , HeLa Cells , Humans , Immunoprecipitation , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins , Microscopy, Electron , Microscopy, Fluorescence , Poly-ADP-Ribose Binding Proteins , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/metabolism , Time-Lapse Imaging , Ubiquitin-Protein Ligases , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL