Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2985, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35624105

ABSTRACT

The speed and efficiency of data collection and image processing in cryo-electron microscopy have increased over the last decade. However, cryo specimen preparation techniques have lagged and faster, more reproducible specimen preparation devices are needed. Here, we present a vitrification device with highly automated sample handling, requiring only limited user interaction. Moreover, the device allows inspection of thin films using light microscopy, since the excess liquid is removed through suction by tubes, not blotting paper. In combination with dew-point control, this enables thin film preparation in a controlled and reproducible manner. The advantage is that the quality of the prepared cryo specimen is characterized before electron microscopy data acquisition. The practicality and performance of the device are illustrated with experimental results obtained by vitrification of protein suspensions, lipid vesicles, bacterial and human cells, followed by imaged using single particle analysis, cryo-electron tomography, and cryo correlated light and electron microscopy.


Subject(s)
Electron Microscope Tomography , Vitrification , Cryoelectron Microscopy/methods , Humans , Proteins , Suction
2.
Commun Biol ; 5(1): 29, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017666

ABSTRACT

Cryo-electron microscopy has become an essential tool to understand structure and function of biological samples. Especially for pathogens, such as disease-causing bacteria and viruses, insights gained by cryo-EM can aid in developing cures. However, due to the biosafety restrictions of pathogens, samples are often treated by chemical fixation to render the pathogen inert, affecting the ultrastructure of the sample. Alternatively, researchers use in vitro or ex vivo models, which are non-pathogenic but lack the complexity of the pathogen of interest. Here we show that ultraviolet-C (UVC) radiation applied at cryogenic temperatures can be used to eliminate or dramatically reduce the infectivity of Vibrio cholerae and the bacterial virus, the ICP1 bacteriophage. We show no discernable structural impact of this treatment of either sample using two cryo-EM methods: cryo-electron tomography followed by sub-tomogram averaging, and single particle analysis (SPA). Additionally, we applied the UVC irradiation to the protein apoferritin (ApoF), which is a widely used test sample for high-resolution SPA studies. The UVC-treated ApoF sample resulted in a 2.1 Å structure indistinguishable from an untreated published map. This research demonstrates that UVC treatment is an effective and inexpensive addition to the cryo-EM sample preparation toolbox.


Subject(s)
Bacteria , Cryoelectron Microscopy , Ultraviolet Rays , Viruses , Bacteria/pathogenicity , Bacteria/radiation effects , Chemotaxis/radiation effects , Vibrio cholerae/pathogenicity , Vibrio cholerae/radiation effects , Viruses/pathogenicity , Viruses/radiation effects
3.
Methods Mol Biol ; 2305: 229-256, 2021.
Article in English | MEDLINE | ID: mdl-33950393

ABSTRACT

In this chapter, we present an overview of a standard protocol to achieve structure determination at high resolution by Single Particle Analysis cryogenic Electron Microscopy using apoferritin as a standard sample. The purified apoferritin is applied to a glow-discharged support and then flash frozen in liquid ethane. The prepared grids are loaded into the electron microscope and checked for particle spreading and ice thickness. The microscope alignments are performed and the data collection session is setup for an overnight data collection. The collected movies containing two-dimensional images of the apoferritin sample are then processed to obtain a high-resolution three-dimensional reconstruction.


Subject(s)
Apoferritins/chemistry , Cryoelectron Microscopy/instrumentation , Cryoelectron Microscopy/methods , Specimen Handling/methods , Animals , Apoferritins/ultrastructure , Equidae , Freezing , Imaging, Three-Dimensional , Workflow
4.
Nat Commun ; 12(1): 244, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431842

ABSTRACT

The trimeric spike (S) protein of SARS-CoV-2 is the primary focus of most vaccine design and development efforts. Due to intrinsic instability typical of class I fusion proteins, S tends to prematurely refold to the post-fusion conformation, compromising immunogenic properties and prefusion trimer yields. To support ongoing vaccine development efforts, we report the structure-based design of soluble S trimers with increased yields and stabilities, based on introduction of single point mutations and disulfide-bridges. We identify regions critical for stability: the heptad repeat region 1, the SD1 domain and position 614 in SD2. We combine a minimal selection of mostly interprotomeric mutations to create a stable S-closed variant with a 6.4-fold higher expression than the parental construct while no longer containing a heterologous trimerization domain. The cryo-EM structure reveals a correctly folded, predominantly closed pre-fusion conformation. Highly stable and well producing S protein and the increased understanding of S protein structure will support vaccine development and serological diagnostics.


Subject(s)
SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Cryoelectron Microscopy , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Stability , SARS-CoV-2/chemistry , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/genetics
5.
DNA Repair (Amst) ; 85: 102746, 2020 01.
Article in English | MEDLINE | ID: mdl-31739207

ABSTRACT

Nucleotide excision repair (NER) is a DNA repair pathway present in all domains of life. In bacteria, UvrA protein localizes the DNA lesion, followed by verification by UvrB helicase and excision by UvrC double nuclease. UvrA senses deformations and flexibility of the DNA duplex without precisely localizing the lesion in the damaged strand, an element essential for proper NER. Using a combination of techniques, we elucidate the mechanism of the damage verification step in bacterial NER. UvrA dimer recruits two UvrB molecules to its two sides. Each of the two UvrB molecules clamps a different DNA strand using its ß-hairpin element. Both UvrB molecules then translocate to the lesion, and UvrA dissociates. The UvrB molecule that clamps the damaged strand gets stalled at the lesion to recruit UvrC. This mechanism allows UvrB to verify the DNA damage and identify its precise location triggering subsequent steps in the NER pathway.


Subject(s)
Bacteria/genetics , DNA Helicases/chemistry , DNA Helicases/metabolism , Adenosine Triphosphatases/metabolism , Bacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA Damage , DNA Repair , Endodeoxyribonucleases/metabolism , Models, Molecular , Protein Conformation
6.
Nat Commun ; 10(1): 4189, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519882

ABSTRACT

Retroviral integrase can efficiently utilise nucleosomes for insertion of the reverse-transcribed viral DNA. In face of the structural constraints imposed by the nucleosomal structure, integrase gains access to the scissile phosphodiester bonds by lifting DNA off the histone octamer at the site of integration. To clarify the mechanism of DNA looping by integrase, we determined a 3.9 Å resolution structure of the prototype foamy virus intasome engaged with a nucleosome core particle. The structural data along with complementary single-molecule Förster resonance energy transfer measurements reveal twisting and sliding of the nucleosomal DNA arm proximal to the integration site. Sliding the nucleosomal DNA by approximately two base pairs along the histone octamer accommodates the necessary DNA lifting from the histone H2A-H2B subunits to allow engagement with the intasome. Thus, retroviral integration into nucleosomes involves the looping-and-sliding mechanism for nucleosomal DNA repositioning, bearing unexpected similarities to chromatin remodelers.


Subject(s)
Cryoelectron Microscopy/methods , DNA/chemistry , Histones/chemistry , Nucleosomes/metabolism , DNA/ultrastructure , Fluorescence Resonance Energy Transfer , Histones/ultrastructure , Humans , Nucleoproteins/chemistry , Nucleoproteins/ultrastructure , Nucleosomes/ultrastructure , Protein Structure, Quaternary
7.
Biomolecules ; 9(8)2019 08 14.
Article in English | MEDLINE | ID: mdl-31416261

ABSTRACT

With the recent technological and computational advancements, structural biology has begun to tackle more and more difficult questions, including complex biochemical pathways and transient interactions among macromolecules. This has demonstrated that, to approach the complexity of biology, one single technique is largely insufficient and unable to yield thorough answers, whereas integrated approaches have been more and more adopted with successful results. Traditional structural techniques (X-ray crystallography and Nuclear Magnetic Resonance (NMR)) and the emerging ones (cryo-electron microscopy (cryo-EM), Small Angle X-ray Scattering (SAXS)), together with molecular modeling, have pros and cons which very nicely complement one another. In this review, three examples of synergistic approaches chosen from our previous research will be revisited. The first shows how the joint use of both solution and solid-state NMR (SSNMR), X-ray crystallography, and cryo-EM is crucial to elucidate the structure of polyethylene glycol (PEG)ylated asparaginase, which would not be obtainable through any of the techniques taken alone. The second deals with the integrated use of NMR, X-ray crystallography, and SAXS in order to elucidate the catalytic mechanism of an enzyme that is based on the flexibility of the enzyme itself. The third one shows how it is possible to put together experimental data from X-ray crystallography and NMR restraints in order to refine a protein model in order to obtain a structure which simultaneously satisfies both experimental datasets and is therefore closer to the 'real structure'.


Subject(s)
Asparaginase/chemistry , Polyethylene Glycols/chemistry , Asparaginase/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Polyethylene Glycols/metabolism , Scattering, Small Angle , X-Ray Diffraction
8.
J Struct Biol ; 199(3): 225-236, 2017 09.
Article in English | MEDLINE | ID: mdl-28827185

ABSTRACT

This paper provides an overview of the discussion and presentations from the Workshop on the Management of Large CryoEM Facilities held at the New York Structural Biology Center, New York, NY on February 6-7, 2017. A major objective of the workshop was to discuss best practices for managing cryoEM facilities. The discussions were largely focused on supporting single-particle methods for cryoEM and topics included: user access, assessing projects, workflow, sample handling, microscopy, data management and processing, and user training.


Subject(s)
Cryoelectron Microscopy , Research/organization & administration , Cryoelectron Microscopy/instrumentation , Workflow
9.
Nat Commun ; 8: 15720, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28643783

ABSTRACT

ORC, Cdc6 and Cdt1 act together to load hexameric MCM, the motor of the eukaryotic replicative helicase, into double hexamers at replication origins. Here we show that Cdt1 interacts with MCM subunits Mcm2, 4 and 6, which both destabilizes the Mcm2-5 interface and inhibits MCM ATPase activity. Using X-ray crystallography, we show that Cdt1 contains two winged-helix domains in the C-terminal half of the protein and a catalytically inactive dioxygenase-related N-terminal domain, which is important for MCM loading, but not for subsequent replication. We used these structures together with single-particle electron microscopy to generate three-dimensional models of MCM complexes. These show that Cdt1 stabilizes MCM in a left-handed spiral open at the Mcm2-5 gate. We propose that Cdt1 acts as a brace, holding MCM open for DNA entry and bound to ATP until ORC-Cdc6 triggers ATP hydrolysis by MCM, promoting both Cdt1 ejection and MCM ring closure.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Minichromosome Maintenance Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/chemistry , Cell Cycle Proteins/genetics , Cross-Linking Reagents/chemistry , Crystallography, X-Ray , DNA Replication , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , Hydrolysis , Microscopy, Electron , Models, Molecular , Origin Recognition Complex/metabolism , Protein Conformation , Protein Domains , Replication Origin , Saccharomyces cerevisiae Proteins/genetics
10.
Proc Natl Acad Sci U S A ; 114(16): 4141-4146, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28373564

ABSTRACT

The replisome unwinds and synthesizes DNA for genome duplication. In eukaryotes, the Cdc45-MCM-GINS (CMG) helicase and the leading-strand polymerase, Pol epsilon, form a stable assembly. The mechanism for coupling DNA unwinding with synthesis is starting to be elucidated, however the architecture and dynamics of the replication fork remain only partially understood, preventing a molecular understanding of chromosome replication. To address this issue, we conducted a systematic single-particle EM study on multiple permutations of the reconstituted CMG-Pol epsilon assembly. Pol epsilon contains two flexibly tethered lobes. The noncatalytic lobe is anchored to the motor of the helicase, whereas the polymerization domain extends toward the side of the helicase. We observe two alternate configurations of the DNA synthesis domain in the CMG-bound Pol epsilon. We propose that this conformational switch might control DNA template engagement and release, modulating replisome progression.


Subject(s)
DNA Helicases/metabolism , DNA Polymerase II/metabolism , DNA Replication , Eukaryotic Cells/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , DNA Helicases/genetics , DNA Polymerase II/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
11.
Cell Rep ; 18(3): 611-623, 2017 01 17.
Article in English | MEDLINE | ID: mdl-27986592

ABSTRACT

Activation of the main DNA interstrand crosslink repair pathway in higher eukaryotes requires mono-ubiquitination of FANCI and FANCD2 by FANCL, the E3 ligase subunit of the Fanconi anemia core complex. FANCI and FANCD2 form a stable complex; however, the molecular basis of their ubiquitination is ill defined. FANCD2 mono-ubiquitination by FANCL is stimulated by the presence of the FANCB and FAAP100 core complex components, through an unknown mechanism. How FANCI mono-ubiquitination is achieved remains unclear. Here, we use structural electron microscopy, combined with crosslink-coupled mass spectrometry, to find that FANCB, FANCL, and FAAP100 form a dimer of trimers, containing two FANCL molecules that are ideally poised to target both FANCI and FANCD2 for mono-ubiquitination. The FANCC-FANCE-FANCF subunits bridge between FANCB-FANCL-FAAP100 and the FANCI-FANCD2 substrate. A transient interaction with FANCC-FANCE-FANCF alters the FANCI-FANCD2 configuration, stabilizing the dimerization interface. Our data provide a model to explain how equivalent mono-ubiquitination of FANCI and FANCD2 occurs.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Amino Acid Sequence , Chromatography, High Pressure Liquid , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Fanconi Anemia Complementation Group D2 Protein/chemistry , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Mass Spectrometry , Microscopy, Electron , Protein Multimerization , Protein Structure, Tertiary , Substrate Specificity , Ubiquitination
12.
PLoS Pathog ; 12(9): e1005835, 2016 09.
Article in English | MEDLINE | ID: mdl-27606840

ABSTRACT

The structure of the infectious prion protein (PrPSc), which is responsible for Creutzfeldt-Jakob disease in humans and bovine spongiform encephalopathy, has escaped all attempts at elucidation due to its insolubility and propensity to aggregate. PrPSc replicates by converting the non-infectious, cellular prion protein (PrPC) into the misfolded, infectious conformer through an unknown mechanism. PrPSc and its N-terminally truncated variant, PrP 27-30, aggregate into amorphous aggregates, 2D crystals, and amyloid fibrils. The structure of these infectious conformers is essential to understanding prion replication and the development of structure-based therapeutic interventions. Here we used the repetitive organization inherent to GPI-anchorless PrP 27-30 amyloid fibrils to analyze their structure via electron cryomicroscopy. Fourier-transform analyses of averaged fibril segments indicate a repeating unit of 19.1 Å. 3D reconstructions of these fibrils revealed two distinct protofilaments, and, together with a molecular volume of 18,990 Å3, predicted the height of each PrP 27-30 molecule as ~17.7 Å. Together, the data indicate a four-rung ß-solenoid structure as a key feature for the architecture of infectious mammalian prions. Furthermore, they allow to formulate a molecular mechanism for the replication of prions. Knowledge of the prion structure will provide important insights into the self-propagation mechanisms of protein misfolding.


Subject(s)
Amyloid/ultrastructure , PrPC Proteins/ultrastructure , PrPSc Proteins/ultrastructure , Amyloid/genetics , Animals , Cattle , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Cryoelectron Microscopy , Encephalopathy, Bovine Spongiform/genetics , Encephalopathy, Bovine Spongiform/metabolism , Encephalopathy, Bovine Spongiform/pathology , Humans , PrPC Proteins/genetics , PrPSc Proteins/genetics
13.
Nat Commun ; 7: 10708, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26888060

ABSTRACT

The Cdc45-MCM-GINS (CMG) helicase unwinds DNA during the elongation step of eukaryotic genome duplication and this process depends on the MCM ATPase function. Whether CMG translocation occurs on single- or double-stranded DNA and how ATP hydrolysis drives DNA unwinding remain open questions. Here we use cryo-electron microscopy to describe two subnanometre resolution structures of the CMG helicase trapped on a DNA fork. In the predominant state, the ring-shaped C-terminal ATPase of MCM is compact and contacts single-stranded DNA, via a set of pre-sensor 1 hairpins that spiral around the translocation substrate. In the second state, the ATPase module is relaxed and apparently substrate free, while DNA intimately contacts the downstream amino-terminal tier of the MCM motor ring. These results, supported by single-molecule FRET measurements, lead us to suggest a replication fork unwinding mechanism whereby the N-terminal and AAA+ tiers of the MCM work in concert to translocate on single-stranded DNA.


Subject(s)
DNA Helicases/metabolism , DNA/metabolism , Eukaryota/enzymology , Cryoelectron Microscopy , DNA/genetics , DNA/ultrastructure , DNA Helicases/ultrastructure , DNA Replication , Eukaryota/genetics , Eukaryota/ultrastructure
14.
Nature ; 523(7560): 366-9, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26061770

ABSTRACT

Retroviral integration is catalysed by a tetramer of integrase (IN) assembled on viral DNA ends in a stable complex, known as the intasome. How the intasome interfaces with chromosomal DNA, which exists in the form of nucleosomal arrays, is currently unknown. Here we show that the prototype foamy virus (PFV) intasome is proficient at stable capture of nucleosomes as targets for integration. Single-particle cryo-electron microscopy reveals a multivalent intasome-nucleosome interface involving both gyres of nucleosomal DNA and one H2A-H2B heterodimer. While the histone octamer remains intact, the DNA is lifted from the surface of the H2A-H2B heterodimer to allow integration at strongly preferred superhelix location ±3.5 positions. Amino acid substitutions disrupting these contacts impinge on the ability of the intasome to engage nucleosomes in vitro and redistribute viral integration sites on the genomic scale. Our findings elucidate the molecular basis for nucleosome capture by the viral DNA recombination machinery and the underlying nucleosome plasticity that allows integration.


Subject(s)
Nucleosomes/chemistry , Nucleosomes/virology , Spumavirus/metabolism , Virus Integration , Amino Acid Substitution , Binding Sites/genetics , Cryoelectron Microscopy , DNA/genetics , DNA/metabolism , DNA/ultrastructure , Genome/genetics , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Integrases/metabolism , Models, Molecular , Nucleosomes/genetics , Nucleosomes/ultrastructure , Protein Multimerization , Recombination, Genetic , Spumavirus/chemistry , Spumavirus/genetics , Spumavirus/ultrastructure
15.
Proc Natl Acad Sci U S A ; 112(14): 4286-91, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25831490

ABSTRACT

RecQ helicases are a widely conserved family of ATP-dependent motors with diverse roles in nearly every aspect of bacterial and eukaryotic genome maintenance. However, the physical mechanisms by which RecQ helicases recognize and process specific DNA replication and repair intermediates are largely unknown. Here, we solved crystal structures of the human RECQ1 helicase in complexes with tailed-duplex DNA and ssDNA. The structures map the interactions of the ssDNA tail and the branch point along the helicase and Zn-binding domains, which, together with reported structures of other helicases, define the catalytic stages of helicase action. We also identify a strand-separating pin, which (uniquely in RECQ1) is buttressed by the protein dimer interface. A duplex DNA-binding surface on the C-terminal domain is shown to play a role in DNA unwinding, strand annealing, and Holliday junction (HJ) branch migration. We have combined EM and analytical ultracentrifugation approaches to show that RECQ1 can form what appears to be a flat, homotetrameric complex and propose that RECQ1 tetramers are involved in HJ recognition. This tetrameric arrangement suggests a platform for coordinated activity at the advancing and receding duplexes of an HJ during branch migration.


Subject(s)
DNA Helicases/chemistry , DNA/chemistry , RecQ Helicases/chemistry , Animals , Chromatography, Gel , Crystallization , Crystallography, X-Ray , DNA, Cruciform/physiology , DNA, Single-Stranded/chemistry , Escherichia coli/metabolism , Humans , Insecta , Molecular Conformation , Nucleic Acid Denaturation , Nucleotides/chemistry , Protein Binding , Protein Structure, Tertiary , Zinc/chemistry
16.
J Biol Chem ; 290(3): 1522-35, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25411244

ABSTRACT

The acetylcholinesterase found in the venom of Bungarus fasciatus (BfAChE) is produced as a soluble, non-amphiphilic monomer with a canonical catalytic domain but a distinct C terminus compared with the other vertebrate enzymes. Moreover, the peripheral anionic site of BfAChE, a surface site located at the active site gorge entrance, bears two substitutions altering sensitivity to cationic inhibitors. Antibody Elec410, generated against Electrophorus electricus acetylcholinesterase (EeAChE), inhibits EeAChE and BfAChE by binding to their peripheral sites. However, both complexes retain significant residual catalytic activity, suggesting incomplete gorge occlusion by bound antibody and/or high frequency back door opening. To explore a novel acetylcholinesterase species, ascertain the molecular bases of inhibition by Elec410, and document the determinants and mechanisms for back door opening, we solved a 2.7-Å resolution crystal structure of natural BfAChE in complex with antibody fragment Fab410. Crystalline BfAChE forms the canonical dimer found in all acetylcholinesterase structures. Equally represented open and closed states of a back door channel, associated with alternate positions of a tyrosine phenol ring at the active site base, coexist in each subunit. At the BfAChE molecular surface, Fab410 is seated on the long Ω-loop between two N-glycan chains and partially occludes the gorge entrance, a position that fully reflects the available mutagenesis and biochemical data. Experimentally based flexible molecular docking supports a similar Fab410 binding mode onto the EeAChE antigen. These data document the molecular and dynamic peculiarities of BfAChE with high frequency back door opening, and the mode of action of Elec410 as one of the largest peptidic inhibitors targeting the acetylcholinesterase peripheral site.


Subject(s)
Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Immunoglobulin Fragments/chemistry , Snake Venoms/enzymology , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antigens/chemistry , Catalysis , Catalytic Domain , Crystallography, X-Ray , Humans , Molecular Sequence Data , Phenol/chemistry , Polysaccharides/chemistry , Protein Binding , Protein Conformation , Protein Multimerization , Sequence Homology, Amino Acid , Solvents/chemistry
17.
Elife ; 3: e03273, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25117490

ABSTRACT

The Cdc45/Mcm2-7/GINS (CMG) helicase separates DNA strands during replication in eukaryotes. How the CMG is assembled and engages DNA substrates remains unclear. Using electron microscopy, we have determined the structure of the CMG in the presence of ATPγS and a DNA duplex bearing a 3' single-stranded tail. The structure shows that the MCM subunits of the CMG bind preferentially to single-stranded DNA, establishes the polarity by which DNA enters into the Mcm2-7 pore, and explains how Cdc45 helps prevent DNA from dissociating from the helicase. The Mcm2-7 subcomplex forms a cracked-ring, right-handed spiral when DNA and nucleotide are bound, revealing unexpected congruencies between the CMG and both bacterial DnaB helicases and the AAA+ motor of the eukaryotic proteasome. The existence of a subpopulation of dimeric CMGs establishes the subunit register of Mcm2-7 double hexamers and together with the spiral form highlights how Mcm2-7 transitions through different conformational and assembly states as it matures into a functional helicase.


Subject(s)
Cell Cycle Proteins/chemistry , Chromosomal Proteins, Non-Histone/chemistry , DNA-Binding Proteins/chemistry , Drosophila Proteins/chemistry , Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/metabolism , Multiprotein Complexes/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA/chemistry , DNA/metabolism , DNA Replication , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Eukaryotic Cells/metabolism , Microscopy, Electron , Models, Molecular , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA Splicing Factors , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism
18.
Nature ; 510(7504): 293-297, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24805245

ABSTRACT

Efficient duplication of the genome requires the concerted action of helicase and DNA polymerases at replication forks to avoid stalling of the replication machinery and consequent genomic instability. In eukaryotes, the physical coupling between helicase and DNA polymerases remains poorly understood. Here we define the molecular mechanism by which the yeast Ctf4 protein links the Cdc45-MCM-GINS (CMG) DNA helicase to DNA polymerase α (Pol α) within the replisome. We use X-ray crystallography and electron microscopy to show that Ctf4 self-associates in a constitutive disk-shaped trimer. Trimerization depends on a ß-propeller domain in the carboxy-terminal half of the protein, which is fused to a helical extension that protrudes from one face of the trimeric disk. Critically, Pol α and the CMG helicase share a common mechanism of interaction with Ctf4. We show that the amino-terminal tails of the catalytic subunit of Pol α and the Sld5 subunit of GINS contain a conserved Ctf4-binding motif that docks onto the exposed helical extension of a Ctf4 protomer within the trimer. Accordingly, we demonstrate that one Ctf4 trimer can support binding of up to three partner proteins, including the simultaneous association with both Pol α and GINS. Our findings indicate that Ctf4 can couple two molecules of Pol α to one CMG helicase within the replisome, providing a new model for lagging-strand synthesis in eukaryotes that resembles the emerging model for the simpler replisome of Escherichia coli. The ability of Ctf4 to act as a platform for multivalent interactions illustrates a mechanism for the concurrent recruitment of factors that act together at the fork.


Subject(s)
DNA Helicases/metabolism , DNA Polymerase I/metabolism , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Protein Multimerization , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , DNA Helicases/chemistry , DNA Helicases/ultrastructure , DNA Polymerase I/chemistry , DNA Polymerase I/ultrastructure , DNA-Binding Proteins/ultrastructure , Microscopy, Electron , Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure
19.
PLoS One ; 8(10): e77226, 2013.
Article in English | MEDLINE | ID: mdl-24146971

ABSTRACT

The inhibition properties and target sites of monoclonal antibodies (mAbs) Elec403, Elec408 and Elec410, generated against Electrophorus electricus acetylcholinesterase (AChE), have been defined previously using biochemical and mutagenesis approaches. Elec403 and Elec410, which bind competitively with each other and with the peptidic toxin inhibitor fasciculin, are directed toward distinctive albeit overlapping epitopes located at the AChE peripheral anionic site, which surrounds the entrance of the active site gorge. Elec408, which is not competitive with the other two mAbs nor fasciculin, targets a second epitope located in the backdoor region, distant from the gorge entrance. To characterize the molecular determinants dictating their binding site specificity, we cloned and sequenced the mAbs; generated antigen-binding fragments (Fab) retaining the parental inhibition properties; and explored their structure-function relationships using complementary x-ray crystallography, homology modeling and flexible docking approaches. Hypermutation of one Elec403 complementarity-determining region suggests occurrence of antigen-driven selection towards recognition of the AChE peripheral site. Comparative analysis of the 1.9Å-resolution structure of Fab408 and of theoretical models of its Fab403 and Fab410 congeners evidences distinctive surface topographies and anisotropic repartitions of charges, consistent with their respective target sites and inhibition properties. Finally, a validated, data-driven docking model of the Fab403-AChE complex suggests a mode of binding at the PAS that fully correlates with the functional data. This comprehensive study documents the molecular peculiarities of Fab403 and Fab410, as the largest peptidic inhibitors directed towards the peripheral site, and those of Fab408, as the first inhibitor directed toward the backdoor region of an AChE and a unique template for the design of new, specific modulators of AChE catalysis.


Subject(s)
Acetylcholinesterase/metabolism , Acetylcholinesterase/chemistry , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Binding Sites , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacology , Cloning, Molecular , Complementarity Determining Regions , Enzyme Activation , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Models, Molecular , Molecular Docking Simulation , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Alignment , Sequence Analysis, DNA
20.
J Biol Chem ; 288(7): 4772-81, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23283973

ABSTRACT

Prion diseases are fatal neurodegenerative disorders associated with the polymerization of the cellular form of prion protein (PrP(C)) into an amyloidogenic ß-sheet infectious form (PrP(Sc)). The sequence of host PrP is the major determinant of host prion disease susceptibility. In mice, the presence of allele a (Prnp(a), encoding the polymorphism Leu-108/Thr-189) or b (Prnp(b), Phe-108/Val-189) is associated with short or long incubation times, respectively, following infection with PrP(Sc). The molecular bases linking PrP sequence, infection susceptibility, and convertibility of PrP(C) into PrP(Sc) remain unclear. Here we show that recombinant PrP(a) and PrP(b) aggregate and respond to seeding differently in vitro. Our kinetic studies reveal differences during the nucleation phase of the aggregation process, where PrP(b) exhibits a longer lag phase that cannot be completely eliminated by seeding the reaction with preformed fibrils. Additionally, PrP(b) is more prone to propagate features of the seeds, as demonstrated by conformational stability and electron microscopy studies of the formed fibrils. We propose a model of polymerization to explain how the polymorphisms at positions 108 and 189 produce the phenotypes seen in vivo. This model also provides insight into phenomena such as species barrier and prion strain generation, two phenomena also influenced by the primary structure of PrP.


Subject(s)
Phenylalanine/genetics , Polymorphism, Genetic , Prion Diseases/metabolism , Prions/genetics , Valine/genetics , Alleles , Animals , Benzothiazoles , Circular Dichroism , Dose-Response Relationship, Drug , Kinetics , Mice , Microscopy, Electron/methods , Polymers/chemistry , Prion Diseases/genetics , Protein Conformation , Recombinant Proteins/metabolism , Thiazoles/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...