Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Nature ; 612(7938): 162-169, 2022 12.
Article in English | MEDLINE | ID: mdl-36418402

ABSTRACT

The poly-ADP-ribosyltransferase tankyrase (TNKS, TNKS2) controls a wide range of disease-relevant cellular processes, including WNT-ß-catenin signalling, telomere length maintenance, Hippo signalling, DNA damage repair and glucose homeostasis1,2. This has incentivized the development of tankyrase inhibitors. Notwithstanding, our knowledge of the mechanisms that control tankyrase activity has remained limited. Both catalytic and non-catalytic functions of tankyrase depend on its filamentous polymerization3-5. Here we report the cryo-electron microscopy reconstruction of a filament formed by a minimal active unit of tankyrase, comprising the polymerizing sterile alpha motif (SAM) domain and its adjacent catalytic domain. The SAM domain forms a novel antiparallel double helix, positioning the protruding catalytic domains for recurring head-to-head and tail-to-tail interactions. The head interactions are highly conserved among tankyrases and induce an allosteric switch in the active site within the catalytic domain to promote catalysis. Although the tail interactions have a limited effect on catalysis, they are essential to tankyrase function in WNT-ß-catenin signalling. This work reveals a novel SAM domain polymerization mode, illustrates how supramolecular assembly controls catalytic and non-catalytic functions, provides important structural insights into the regulation of a non-DNA-dependent poly-ADP-ribosyltransferase and will guide future efforts to modulate tankyrase and decipher its contribution to disease mechanisms.


Subject(s)
Biocatalysis , Cryoelectron Microscopy , Polymerization , Tankyrases , beta Catenin , Tankyrases/chemistry , Tankyrases/metabolism , Tankyrases/ultrastructure , Enzyme Activation , Catalytic Domain , Wnt Signaling Pathway , Amino Acid Motifs
2.
Mol Cell ; 79(1): 99-114.e9, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32445620

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes are essential for genome organization from bacteria to humans, but their mechanisms of action remain poorly understood. Here, we characterize human SMC complexes condensin I and II and unveil the architecture of the human condensin II complex, revealing two putative DNA-entrapment sites. Using single-molecule imaging, we demonstrate that both condensin I and II exhibit ATP-dependent motor activity and promote extensive and reversible compaction of double-stranded DNA. Nucleosomes are incorporated into DNA loops during compaction without being displaced from the DNA, indicating that condensin complexes can readily act upon nucleosome-bound DNA molecules. These observations shed light on critical processes involved in genome organization in human cells.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nucleosomes/metabolism , Adenosine Triphosphatases/genetics , DNA-Binding Proteins/genetics , Humans , Models, Molecular , Multiprotein Complexes/genetics , Protein Binding , Protein Conformation , Single Molecule Imaging/methods
3.
Cell ; 139(4): 719-30, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19896182

ABSTRACT

The licensing of eukaryotic DNA replication origins, which ensures once-per-cell-cycle replication, involves the loading of six related minichromosome maintenance proteins (Mcm2-7) into prereplicative complexes (pre-RCs). Mcm2-7 forms the core of the replicative DNA helicase, which is inactive in the pre-RC. The loading of Mcm2-7 onto DNA requires the origin recognition complex (ORC), Cdc6, and Cdt1, and depends on ATP. We have reconstituted Mcm2-7 loading with purified budding yeast proteins. Using biochemical approaches and electron microscopy, we show that single heptamers of Cdt1*Mcm2-7 are loaded cooperatively and result in association of stable, head-to-head Mcm2-7 double hexamers connected via their N-terminal rings. DNA runs through a central channel in the double hexamer, and, once loaded, Mcm2-7 can slide passively along double-stranded DNA. Our work has significant implications for understanding how eukaryotic DNA replication origins are chosen and licensed, how replisomes assemble during initiation, and how unwinding occurs during DNA replication.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/isolation & purification , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/isolation & purification , DNA Helicases/metabolism , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Minichromosome Maintenance Complex Component 3 , Minichromosome Maintenance Complex Component 4 , Minichromosome Maintenance Complex Component 6 , Minichromosome Maintenance Complex Component 7 , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/isolation & purification , Origin Recognition Complex/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/isolation & purification
4.
Nature ; 553(7688): 301-306, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29345637

ABSTRACT

RNA polymerase (Pol) III transcribes essential non-coding RNAs, including the entire pool of transfer RNAs, the 5S ribosomal RNA and the U6 spliceosomal RNA, and is often deregulated in cancer cells. The initiation of gene transcription by Pol III requires the activity of the transcription factor TFIIIB to form a transcriptionally active Pol III preinitiation complex (PIC). Here we present electron microscopy reconstructions of Pol III PICs at 3.4-4.0 Å and a reconstruction of unbound apo-Pol III at 3.1 Å. TFIIIB fully encircles the DNA and restructures Pol III. In particular, binding of the TFIIIB subunit Bdp1 rearranges the Pol III-specific subunits C37 and C34, thereby promoting DNA opening. The unwound DNA directly contacts both sides of the Pol III cleft. Topologically, the Pol III PIC resembles the Pol II PIC, whereas the Pol I PIC is more divergent. The structures presented unravel the molecular mechanisms underlying the first steps of Pol III transcription and also the general conserved mechanisms of gene transcription initiation.


Subject(s)
RNA Polymerase III/metabolism , RNA Polymerase III/ultrastructure , Transcription Initiation, Genetic , Cryoelectron Microscopy , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , Models, Molecular , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA Polymerase I/chemistry , RNA Polymerase II/chemistry , RNA Polymerase III/chemistry , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Templates, Genetic , Transcription Factor TFIIIB/chemistry , Transcription Factor TFIIIB/metabolism , Transcription Factor TFIIIB/ultrastructure , Transcription Factors, TFII/chemistry
5.
Mol Cell ; 63(3): 498-513, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27494558

ABSTRACT

The poly(ADP-ribose) polymerase (PARP) Tankyrase (TNKS and TNKS2) is paramount to Wnt-ß-catenin signaling and a promising therapeutic target in Wnt-dependent cancers. The pool of active ß-catenin is normally limited by destruction complexes, whose assembly depends on the polymeric master scaffolding protein AXIN. Tankyrase, which poly(ADP-ribosyl)ates and thereby destabilizes AXIN, also can polymerize, but the relevance of these polymers has remained unclear. We report crystal structures of the polymerizing TNKS and TNKS2 sterile alpha motif (SAM) domains, revealing versatile head-to-tail interactions. Biochemical studies informed by these structures demonstrate that polymerization is required for Tankyrase to drive ß-catenin-dependent transcription. We show that the polymeric state supports PARP activity and allows Tankyrase to effectively access destruction complexes through enabling avidity-dependent AXIN binding. This study provides an example for regulated signal transduction in non-membrane-enclosed compartments (signalosomes), and it points to novel potential strategies to inhibit Tankyrase function in oncogenic Wnt signaling.


Subject(s)
Sterile Alpha Motif , Tankyrases/metabolism , Wnt Signaling Pathway , Axin Protein/metabolism , Binding Sites , Caspase Activation and Recruitment Domain , Catalysis , Crystallography , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Models, Molecular , Mutation , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Structure-Activity Relationship , Tankyrases/chemistry , Tankyrases/genetics , Transfection
6.
Nucleic Acids Res ; 49(8): 4534-4549, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33849072

ABSTRACT

The multi-component Smc5/6 complex plays a critical role in the resolution of recombination intermediates formed during mitosis and meiosis, and in the cellular response to replication stress. Using recombinant proteins, we have reconstituted a series of defined Saccharomyces cerevisiae Smc5/6 complexes, visualised them by negative stain electron microscopy, and tested their ability to function as an ATPase. We find that only the six protein 'holo-complex' is capable of turning over ATP and that its activity is significantly increased by the addition of double-stranded DNA to reaction mixes. Furthermore, stimulation is wholly dependent on functional ATP-binding pockets in both Smc5 and Smc6. Importantly, we demonstrate that budding yeast Nse5/6 acts as a negative regulator of Smc5/6 ATPase activity, binding to the head-end of the complex to suppress turnover, irrespective of the DNA-bound status of the complex.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/ultrastructure , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/ultrastructure , DNA/metabolism , Escherichia coli/metabolism , Microscopy, Electron, Transmission , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure
7.
Mol Cell ; 55(5): 666-77, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25087873

ABSTRACT

Loading of the six related Minichromosome Maintenance (MCM) proteins as head-to-head double hexamers during DNA replication origin licensing is crucial for ensuring once-per-cell-cycle DNA replication in eukaryotic cells. Assembly of these prereplicative complexes (pre-RCs) requires the Origin Recognition Complex (ORC), Cdc6, and Cdt1. ORC, Cdc6, and MCM are members of the AAA+ family of ATPases, and pre-RC assembly requires ATP hydrolysis. Here we show that ORC and Cdc6 mutants defective in ATP hydrolysis are competent for origin licensing. However, ATP hydrolysis by Cdc6 is required to release nonproductive licensing intermediates. We show that ATP binding stabilizes the wild-type MCM hexamer. Moreover, by analyzing MCM containing mutant subunits, we show that ATP binding and hydrolysis by MCM are required for Cdt1 release and double hexamer formation. This work alters our view of how ATP is used by licensing factors to assemble pre-RCs.


Subject(s)
Adenosine Triphosphate/metabolism , DNA Replication/physiology , Minichromosome Maintenance Proteins/physiology , Replication Origin , Adenosine Triphosphate/physiology , Amino Acid Sequence , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Hydrolysis , Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/metabolism , Molecular Sequence Data , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , Origin Recognition Complex/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Sequence Alignment
8.
EMBO J ; 33(6): 605-20, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24566989

ABSTRACT

Eukaryotic DNA replication initiates from multiple replication origins. To ensure each origin fires just once per cell cycle, initiation is divided into two biochemically discrete steps: the Mcm2-7 helicase is first loaded into prereplicative complexes (pre-RCs) as an inactive double hexamer by the origin recognition complex (ORC), Cdt1 and Cdc6; the helicase is then activated by a set of "firing factors." Here, we show that plasmids containing pre-RCs assembled with purified proteins support complete and semi-conservative replication in extracts from budding yeast cells overexpressing firing factors. Replication requires cyclin-dependent kinase (CDK) and Dbf4-dependent kinase (DDK). DDK phosphorylation of Mcm2-7 does not by itself promote separation of the double hexamer, but is required for the recruitment of firing factors and replisome components in the extract. Plasmid replication does not require a functional replication origin; however, in the presence of competitor DNA and limiting ORC concentrations, replication becomes origin-dependent in this system. These experiments indicate that Mcm2-7 double hexamers can be precursors of replication and provide insight into the nature of eukaryotic DNA replication origins.


Subject(s)
DNA Replication/physiology , Enzyme Activation/physiology , Minichromosome Maintenance Proteins/metabolism , Multiprotein Complexes/physiology , Replication Origin/physiology , Cell Cycle Proteins/metabolism , Mass Spectrometry , Models, Biological , Models, Molecular , Phosphorylation , Plasmids/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomycetales
9.
Chembiochem ; 17(12): 1107-10, 2016 06 16.
Article in English | MEDLINE | ID: mdl-26992063

ABSTRACT

Nanoscale objects of increasing complexity can be constructed from DNA or RNA. However, the scope of potential applications could be enhanced by expanding beyond the moderate chemical diversity of natural nucleic acids. Here, we explore the construction of nano-objects made entirely from alternative building blocks: synthetic genetic polymers not found in nature, also called xeno nucleic acids (XNAs). Specifically, we describe assembly of 70 kDa tetrahedra elaborated in four different XNA chemistries (2'-fluro-2'-deoxy-ribofuranose nucleic acid (2'F-RNA), 2'-fluoroarabino nucleic acids (FANA), hexitol nucleic acids (HNA), and cyclohexene nucleic acids (CeNA)), as well as mixed designs, and a ∼600 kDa all-FANA octahedron, visualised by electron microscopy. Our results extend the chemical scope for programmable nanostructure assembly, with implications for the design of nano-objects and materials with an expanded range of structural and physicochemical properties, including enhanced biostability.


Subject(s)
Nanostructures/chemistry , Polymers/chemistry , Electrophoretic Mobility Shift Assay , Microscopy, Electron, Transmission , Nucleic Acids/chemistry
10.
EMBO J ; 30(15): 3078-90, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21701561

ABSTRACT

The cytosolic chaperonin CCT is a 1-MDa protein-folding machine essential for eukaryotic life. The CCT interactome shows involvement in folding and assembly of a small range of proteins linked to essential cellular processes such as cytoskeleton assembly and cell-cycle regulation. CCT has a classic chaperonin architecture, with two heterogeneous 8-membered rings stacked back-to-back, enclosing a folding cavity. However, the mechanism by which CCT assists folding is distinct from other chaperonins, with no hydrophobic wall lining a potential Anfinsen cage, and a sequential rather than concerted ATP hydrolysis mechanism. We have solved the crystal structure of yeast CCT in complex with actin at 3.8 Å resolution, revealing the subunit organisation and the location of discrete patches of co-evolving 'signature residues' that mediate specific interactions between CCT and its substrates. The intrinsic asymmetry is revealed by the structural individuality of the CCT subunits, which display unique configurations, substrate binding properties, ATP-binding heterogeneity and subunit-subunit interactions. The location of the evolutionarily conserved N-terminus of Cct5 on the outside of the barrel, confirmed by mutational studies, is unique to eukaryotic cytosolic chaperonins.


Subject(s)
Chaperonin Containing TCP-1/chemistry , Saccharomyces cerevisiae/enzymology , Actins/chemistry , Actins/metabolism , Amino Acid Sequence , Chaperonin Containing TCP-1/metabolism , Crystallography, X-Ray , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/chemistry
11.
PLoS Biol ; 10(2): e1001261, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22347812

ABSTRACT

Active muscles generate substantial mechanical forces by the contraction/relaxation cycle, and, to maintain an ordered state, they require molecular structures of extraordinary stability. These forces are sensed and buffered by unusually long and elastic filament proteins with highly repetitive domain arrays. Members of the myomesin protein family function as molecular bridges that connect major filament systems in the central M-band of muscle sarcomeres, which is a central locus of passive stress sensing. To unravel the mechanism of molecular elasticity in such filament-connecting proteins, we have determined the overall architecture of the complete C-terminal immunoglobulin domain array of myomesin by X-ray crystallography, electron microscopy, solution X-ray scattering, and atomic force microscopy. Our data reveal a dimeric tail-to-tail filament structure of about 360 Å in length, which is folded into an irregular superhelical coil arrangement of almost identical α-helix/domain modules. The myomesin filament can be stretched to about 2.5-fold its original length by reversible unfolding of these linkers, a mechanism that to our knowledge has not been observed previously. Our data explain how myomesin could act as a highly elastic ribbon to maintain the overall structural organization of the sarcomeric M-band. In general terms, our data demonstrate how repetitive domain modules such as those found in myomesin could generate highly elastic protein structures in highly organized cell systems such as muscle sarcomeres.


Subject(s)
Elasticity , Muscle Proteins/chemistry , Connectin , Crystallography, X-Ray , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Models, Molecular , Muscle Proteins/ultrastructure , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Sarcomeres/chemistry , Scattering, Small Angle
12.
Front Immunol ; 14: 1204224, 2023.
Article in English | MEDLINE | ID: mdl-37441083

ABSTRACT

Background: Intracellular communication within the tumour is complex and extracellular vesicles (EVs) have been identified as major contributing factors for the cell-to-cell communication in the local and distant tumour environments. Here, we examine the differential effects of breast cancer (BC) subtype-specific patient serum and cell-line derived EVs in the regulation of T cell mediated immune responses. Methods: Ultracentrifugation was used to isolate EVs from sera of 63 BC patients, 15 healthy volunteers and 4 human breast cancer cell lines. Longitudinal blood draws for EV isolation for patients on neoadjuvant chemotherapy was also performed. Characterization of EVs was performed by Nanoparticle Tracking Analysis (NTA), transmission electron microscopy (TEM) and immunoblotting. CD63 staining was performed on a tissue microarray of 218 BC patients. In-house bioinformatics algorithms were utilized for the computation of EV associated expression scores within The Cancer Genome Atlas (TCGA) and correlated with tumour infiltrating lymphocyte (TIL) scores. In vitro stimulation of PBMCs with EVs from serum and cell-line derived EVs was performed and changes in the immune phenotypes characterized by flow cytometry. Cytokine profiles were assessed using a 105-plex immunoassay or IL10 ELISA. Results: Patients with triple negative breast cancers (TNBCs) exhibited the lowest number of EVs in the sera; whilst the highest was detected in ER+HER2+ cancers; reflected also in the higher level of CD63+ vesicles found within the ER+HER2+ local tumour microenvironment. Transcriptomic analysis of the TCGA data identified that samples assigned with lower EV scores had significantly higher abundance of CD4+ memory activated T cells, T follicular cells and CD8 T cells, plasma, and memory B cells; whilst samples with high EV scores were more enriched for anti-inflammatory M2 macrophages and mast cells. A negative correlation between EV expression scores and stromal TIL counts was also observed. In vitro experiments confirmed that circulating EVs within breast cancer subtypes have functionally differing immunomodulatory capabilities, with EVs from patients with the most aggressive breast cancer subtype (TNBCs) demonstrating the most immune-suppressive phenotype (decreased CD3+HLA-DR+ but increased CD3+PD-L1 T cells, increased CD4+CD127-CD25hi T regulatory cells with associated increase in IL10 cytokine production). In depth assessment of the cytokine modulation triggered by the serum/cell line derived exosomes confirmed differential inflammatory cytokine profiles across differing breast cancer subtypes. Studies using the MDA-231 TNBC breast cancer cell-line derived EVs provided further support that TNBC EVs induced the most immunosuppressive response within PBMCs. Discussion: Our study supports further investigations into how tumour derived EVs are a mechanism that cancers can exploit to promote immune suppression; and breast cancer subtypes produce EVs with differing immunomodulatory capabilities. Understanding the intracellular/extracellular pathways implicated in alteration from active to suppressed immune state may provide a promising way forward for restoring immune competence in specific breast cancer patient populations.


Subject(s)
Extracellular Vesicles , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/metabolism , Interleukin-10/metabolism , Cytokines/metabolism , MCF-7 Cells , Extracellular Vesicles/metabolism , Tumor Microenvironment
13.
Nat Commun ; 13(1): 5075, 2022 08 29.
Article in English | MEDLINE | ID: mdl-36038598

ABSTRACT

Genes encoding the core cell cycle machinery are transcriptionally regulated by the MuvB family of protein complexes in a cell cycle-specific manner. Complexes of MuvB with the transcription factors B-MYB and FOXM1 activate mitotic genes during cell proliferation. The mechanisms of transcriptional regulation by these complexes are still poorly characterised. Here, we combine biochemical analysis and in vitro reconstitution, with structural analysis by cryo-electron microscopy and cross-linking mass spectrometry, to functionally examine these complexes. We find that the MuvB:B-MYB complex binds and remodels nucleosomes, thereby exposing nucleosomal DNA. This remodelling activity is supported by B-MYB which directly binds the remodelled DNA. Given the remodelling activity on the nucleosome, we propose that the MuvB:B-MYB complex functions as a pioneer transcription factor complex. In this work, we rationalise prior biochemical and cellular studies and provide a molecular framework of interactions on a protein complex that is key for cell cycle regulation.


Subject(s)
Gene Expression Regulation , Nucleosomes , Transcription Factors/chemistry , Cell Cycle , Cryoelectron Microscopy , DNA , Transcription Factors/metabolism
14.
Dev Cell ; 11(6): 803-16, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17141156

ABSTRACT

We previously reported that p97/p47-assisted membrane fusion is important for the reassembly of organelles at the end of mitosis, but not for their maintenance during interphase. We have now identified a p97 adaptor protein, p37, which forms a complex with p97 in the cytosol and localizes to the Golgi and ER. siRNA experiments revealed that p37 is required for Golgi and ER biogenesis. Injection of anti-p37 antibodies into cells at different cell cycle stages showed that p37 plays an important role in both Golgi and ER maintenance during interphase as well as in their reassembly at the end of mitosis. In an in vitro Golgi reassembly assay, the p97/p37 complex has membrane fusion activity. In contrast to the p97/p47 pathway, this pathway requires p115-GM130 tethering and SNARE GS15, but not syntaxin5. Interestingly, although VCIP135 is also required, its deubiquitinating activity is unnecessary for p97/p37-mediated activities.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Mitosis/physiology , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Cell Cycle , Cloning, Molecular , Gene Library , HeLa Cells , Humans , Immunoprecipitation , Membrane Fusion , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Qa-SNARE Proteins/metabolism , RNA, Small Interfering/pharmacology , Saccharomyces cerevisiae , Sequence Homology, Amino Acid , Two-Hybrid System Techniques , Ubiquitin/metabolism , Vesicular Transport Proteins/metabolism
15.
Nat Commun ; 12(1): 6992, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34848735

ABSTRACT

Retrotransposons are endogenous elements that have the ability to mobilise their DNA between different locations in the host genome. The Ty3 retrotransposon integrates with an exquisite specificity in a narrow window upstream of RNA Polymerase (Pol) III-transcribed genes, representing a paradigm for harmless targeted integration. Here we present the cryo-EM reconstruction at 4.0 Å of an active Ty3 strand transfer complex bound to TFIIIB transcription factor and a tRNA gene. The structure unravels the molecular mechanisms underlying Ty3 targeting specificity at Pol III-transcribed genes and sheds light into the architecture of retrotransposon machinery during integration. Ty3 intasome contacts a region of TBP, a subunit of TFIIIB, which is blocked by NC2 transcription regulator in RNA Pol II-transcribed genes. A newly-identified chromodomain on Ty3 integrase interacts with TFIIIB and the tRNA gene, defining with extreme precision the integration site position.


Subject(s)
RNA Polymerase III/chemistry , RNA-Directed DNA Polymerase/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Genes, Fungal , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , RNA, Transfer/genetics , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Retroelements , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIIIB/metabolism
16.
PLoS Biol ; 5(2): e20, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17227144

ABSTRACT

RecQ helicases are essential for the maintenance of chromosome stability. In addition to DNA unwinding, some RecQ enzymes have an intrinsic DNA strand annealing activity. The function of this dual enzymatic activity and the mechanism that regulates it is, however, unknown. Here, we describe two quaternary forms of the human RECQ1 helicase, higher-order oligomers consistent with pentamers or hexamers, and smaller oligomers consistent with monomers or dimers. Size exclusion chromatography and transmission electron microscopy show that the equilibrium between the two assembly states is affected by single-stranded DNA (ssDNA) and ATP binding, where ATP or ATPgammaS favors the smaller oligomeric form. Our three-dimensional electron microscopy reconstructions of human RECQ1 reveal a complex cage-like structure of approximately 120 A x 130 A with a central pore. This oligomeric structure is stabilized under conditions in which RECQ1 is proficient in strand annealing. In contrast, competition experiments with the ATPase-deficient K119R and E220Q mutants indicate that RECQ1 monomers, or tight binding dimers, are required for DNA unwinding. Collectively, our findings suggest that higher-order oligomers are associated with DNA strand annealing, and lower-order oligomers with DNA unwinding.


Subject(s)
DNA/metabolism , Protein Structure, Quaternary , RecQ Helicases/metabolism , Binding, Competitive , Chromatography, Gel , Humans , Microscopy, Electron
17.
Nat Commun ; 11(1): 6409, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33335104

ABSTRACT

In eukaryotes, RNA Polymerase (Pol) III is specialized for the transcription of tRNAs and other short, untranslated RNAs. Pol III is a determinant of cellular growth and lifespan across eukaryotes. Upregulation of Pol III transcription is observed in cancer and causative Pol III mutations have been described in neurodevelopmental disorders and hypersensitivity to viral infection. Here, we report a cryo-EM reconstruction at 4.0 Å of human Pol III, allowing mapping and rationalization of reported genetic mutations. Mutations causing neurodevelopmental defects cluster in hotspots affecting Pol III stability and/or biogenesis, whereas mutations affecting viral sensing are located in proximity to DNA binding regions, suggesting an impairment of Pol III cytosolic viral DNA-sensing. Integrating x-ray crystallography and SAXS, we also describe the structure of the higher eukaryote specific RPC5 C-terminal extension. Surprisingly, experiments in living cells highlight a role for this module in the assembly and stability of human Pol III.


Subject(s)
RNA Polymerase III/chemistry , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/genetics , Enzyme Stability , HeLa Cells , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Subunits , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Scattering, Small Angle , X-Ray Diffraction
18.
Nat Commun ; 11(1): 1120, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111838

ABSTRACT

The structure-specific endonuclease XPF-ERCC1 participates in multiple DNA damage repair pathways including nucleotide excision repair (NER) and inter-strand crosslink repair (ICLR). How XPF-ERCC1 is catalytically activated by DNA junction substrates is not currently understood. Here we report cryo-electron microscopy structures of both DNA-free and DNA-bound human XPF-ERCC1. DNA-free XPF-ERCC1 adopts an auto-inhibited conformation in which the XPF helical domain masks the ERCC1 (HhH)2 domain and restricts access to the XPF catalytic site. DNA junction engagement releases the ERCC1 (HhH)2 domain to couple with the XPF-ERCC1 nuclease/nuclease-like domains. Structure-function data indicate xeroderma pigmentosum patient mutations frequently compromise the structural integrity of XPF-ERCC1. Fanconi anaemia patient mutations in XPF often display substantial in-vitro activity but are resistant to activation by ICLR recruitment factor SLX4. Our data provide insights into XPF-ERCC1 architecture and catalytic activation.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Endonucleases/chemistry , Endonucleases/metabolism , Binding Sites , Cryoelectron Microscopy , DNA-Binding Proteins/genetics , Endonucleases/genetics , Fanconi Anemia/enzymology , Fanconi Anemia/genetics , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Multimerization , Structure-Activity Relationship , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics
19.
Nat Commun ; 10(1): 3814, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31444342

ABSTRACT

Cullin-Ring E3 Ligases (CRLs) regulate a multitude of cellular pathways through specific substrate receptors. The COP9 signalosome (CSN) deactivates CRLs by removing NEDD8 from activated Cullins. Here we present structures of the neddylated and deneddylated CSN-CRL2 complexes by combining single-particle cryo-electron microscopy (cryo-EM) with chemical cross-linking mass spectrometry (XL-MS). These structures suggest a conserved mechanism of CSN activation, consisting of conformational clamping of the CRL2 substrate by CSN2/CSN4, release of the catalytic CSN5/CSN6 heterodimer and finally activation of the CSN5 deneddylation machinery. Using hydrogen-deuterium exchange (HDX)-MS we show that CRL2 activates CSN5/CSN6 in a neddylation-independent manner. The presence of NEDD8 is required to activate the CSN5 active site. Overall, by synergising cryo-EM with MS, we identify sensory regions of the CSN that mediate its stepwise activation and provide a framework for understanding the regulatory mechanism of other Cullin family members.


Subject(s)
COP9 Signalosome Complex/ultrastructure , NEDD8 Protein/ultrastructure , Peptide Hydrolases/ultrastructure , Ubiquitin-Protein Ligases/ultrastructure , Adaptor Proteins, Signal Transducing/isolation & purification , Adaptor Proteins, Signal Transducing/metabolism , Animals , COP9 Signalosome Complex/isolation & purification , COP9 Signalosome Complex/metabolism , Cryoelectron Microscopy , Intracellular Signaling Peptides and Proteins/isolation & purification , Intracellular Signaling Peptides and Proteins/metabolism , Mass Spectrometry , NEDD8 Protein/isolation & purification , NEDD8 Protein/metabolism , Peptide Hydrolases/isolation & purification , Peptide Hydrolases/metabolism , Protein Processing, Post-Translational , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Sf9 Cells , Ubiquitin-Protein Ligases/isolation & purification , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL