Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Plants ; 9(6): 987-1000, 2023 06.
Article in English | MEDLINE | ID: mdl-37156858

ABSTRACT

In plant cells, translation occurs in three compartments: the cytosol, the plastids and the mitochondria. While the structures of the (prokaryotic-type) ribosomes in plastids and mitochondria are well characterized, high-resolution structures of the eukaryotic 80S ribosomes in the cytosol have been lacking. Here the structure of translating tobacco (Nicotiana tabacum) 80S ribosomes was solved by cryo-electron microscopy with a global resolution of 2.2 Å. The ribosome structure includes two tRNAs, decoded mRNA and the nascent peptide chain, thus providing insights into the molecular underpinnings of the cytosolic translation process in plants. The map displays conserved and plant-specific rRNA modifications and the positions of numerous ionic cofactors, and it uncovers the role of monovalent ions in the decoding centre. The model of the plant 80S ribosome enables broad phylogenetic comparisons that reveal commonalities and differences in the ribosomes of plants and those of other eukaryotes, thus putting our knowledge about eukaryotic translation on a firmer footing.


Subject(s)
RNA, Ribosomal , Ribosomes , Cytosol , RNA, Ribosomal/chemistry , Cryoelectron Microscopy , Phylogeny , Models, Molecular , Ribosomes/chemistry , Plants/genetics , Nicotiana/genetics
2.
Nat Commun ; 13(1): 7641, 2022 12 10.
Article in English | MEDLINE | ID: mdl-36496453

ABSTRACT

Eps15-homology domain containing proteins (EHDs) are eukaryotic, dynamin-related ATPases involved in cellular membrane trafficking. They oligomerize on membranes into filaments that induce membrane tubulation. While EHD crystal structures in open and closed conformations were previously reported, little structural information is available for the membrane-bound oligomeric form. Consequently, mechanistic insights into the membrane remodeling mechanism have remained sparse. Here, by using cryo-electron tomography and subtomogram averaging, we determined structures of nucleotide-bound EHD4 filaments on membrane tubes of various diameters at an average resolution of 7.6 Å. Assembly of EHD4 is mediated via interfaces in the G-domain and the helical domain. The oligomerized EHD4 structure resembles the closed conformation, where the tips of the helical domains protrude into the membrane. The variation in filament geometry and tube radius suggests a spontaneous filament curvature of approximately 1/70 nm-1. Combining the available structural and functional data, we suggest a model for EHD-mediated membrane remodeling.


Subject(s)
Dynamins , Electron Microscope Tomography , Dynamins/metabolism , Adenosine Triphosphatases/metabolism , Cell Membrane/metabolism , Membranes/metabolism , Cryoelectron Microscopy
3.
Science ; 376(6599): 1338-1343, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35709277

ABSTRACT

The elongation of eukaryotic selenoproteins relies on a poorly understood process of interpreting in-frame UGA stop codons as selenocysteine (Sec). We used cryo-electron microscopy to visualize Sec UGA recoding in mammals. A complex between the noncoding Sec-insertion sequence (SECIS), SECIS-binding protein 2 (SBP2), and 40S ribosomal subunit enables Sec-specific elongation factor eEFSec to deliver Sec. eEFSec and SBP2 do not interact directly but rather deploy their carboxyl-terminal domains to engage with the opposite ends of the SECIS. By using its Lys-rich and carboxyl-terminal segments, the ribosomal protein eS31 simultaneously interacts with Sec-specific transfer RNA (tRNASec) and SBP2, which further stabilizes the assembly. eEFSec is indiscriminate toward l-serine and facilitates its misincorporation at Sec UGA codons. Our results support a fundamentally distinct mechanism of Sec UGA recoding in eukaryotes from that in bacteria.


Subject(s)
Codon, Terminator , Peptide Chain Elongation, Translational , RNA-Binding Proteins , Ribosomes , Selenocysteine , Selenoproteins , Codon, Terminator/genetics , Cryoelectron Microscopy , Humans , Peptide Chain Elongation, Translational/genetics , Protein Conformation , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Ribosomes/chemistry , Selenocysteine/chemistry , Selenocysteine/genetics , Selenocysteine/metabolism , Selenoproteins/biosynthesis , Selenoproteins/genetics
4.
Mol Cell ; 81(6): 1200-1215.e9, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33639093

ABSTRACT

Ribosome biogenesis is a fundamental multi-step cellular process that culminates in the formation of ribosomal subunits, whose production and modification are regulated by numerous biogenesis factors. In this study, we analyze physiologic prokaryotic ribosome biogenesis by isolating bona fide pre-50S subunits from an Escherichia coli strain with the biogenesis factor ObgE, affinity tagged at its native gene locus. Our integrative structural approach reveals a network of interacting biogenesis factors consisting of YjgA, RluD, RsfS, and ObgE on the immature pre-50S subunit. In addition, our study provides mechanistic insight into how the GTPase ObgE, in concert with other biogenesis factors, facilitates the maturation of the 50S functional core and reveals both conserved and divergent evolutionary features of ribosome biogenesis between prokaryotes and eukaryotes.


Subject(s)
Escherichia coli Proteins , Evolution, Molecular , Genetic Loci , Hydro-Lyases , Monomeric GTP-Binding Proteins , Ribosome Subunits, Large, Bacterial , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Ribosome Subunits, Large, Bacterial/chemistry , Ribosome Subunits, Large, Bacterial/genetics , Ribosome Subunits, Large, Bacterial/metabolism
5.
Nature ; 578(7795): 467-471, 2020 02.
Article in English | MEDLINE | ID: mdl-31856152

ABSTRACT

Microtubules are dynamic polymers of α- and ß-tubulin and have crucial roles in cell signalling, cell migration, intracellular transport and chromosome segregation1. They assemble de novo from αß-tubulin dimers in an essential process termed microtubule nucleation. Complexes that contain the protein γ-tubulin serve as structural templates for the microtubule nucleation reaction2. In vertebrates, microtubules are nucleated by the 2.2-megadalton γ-tubulin ring complex (γ-TuRC), which comprises γ-tubulin, five related γ-tubulin complex proteins (GCP2-GCP6) and additional factors3. GCP6 is unique among the GCP proteins because it carries an extended insertion domain of unknown function. Our understanding of microtubule formation in cells and tissues is limited by a lack of high-resolution structural information on the γ-TuRC. Here we present the cryo-electron microscopy structure of γ-TuRC from Xenopus laevis at 4.8 Å global resolution, and identify a 14-spoked arrangement of GCP proteins and γ-tubulins in a partially flexible open left-handed spiral with a uniform sequence of GCP variants. By forming specific interactions with other GCP proteins, the GCP6-specific insertion domain acts as a scaffold for the assembly of the γ-TuRC. Unexpectedly, we identify actin as a bona fide structural component of the γ-TuRC with functional relevance in microtubule nucleation. The spiral geometry of γ-TuRC is suboptimal for microtubule nucleation and a controlled conformational rearrangement of the γ-TuRC is required for its activation. Collectively, our cryo-electron microscopy reconstructions provide detailed insights into the molecular organization, assembly and activation mechanism of vertebrate γ-TuRC, and will serve as a framework for the mechanistic understanding of fundamental biological processes associated with microtubule nucleation, such as meiotic and mitotic spindle formation and centriole biogenesis4.


Subject(s)
Cryoelectron Microscopy , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/ultrastructure , Microtubules/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Xenopus , Actins/chemistry , Actins/metabolism , Actins/ultrastructure , Animals , Microtubule-Associated Proteins/metabolism , Microtubules/chemistry , Models, Molecular , Tubulin/chemistry , Tubulin/metabolism , Tubulin/ultrastructure
6.
Cell Rep ; 25(10): 2676-2688.e7, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30517857

ABSTRACT

Translocation moves the tRNA2⋅mRNA module directionally through the ribosome during the elongation phase of protein synthesis. Although translocation is known to entail large conformational changes within both the ribosome and tRNA substrates, the orchestrated events that ensure the speed and fidelity of this critical aspect of the protein synthesis mechanism have not been fully elucidated. Here, we present three high-resolution structures of intermediates of translocation on the mammalian ribosome where, in contrast to bacteria, ribosomal complexes containing the translocase eEF2 and the complete tRNA2⋅mRNA module are trapped by the non-hydrolyzable GTP analog GMPPNP. Consistent with the observed structures, single-molecule imaging revealed that GTP hydrolysis principally facilitates rate-limiting, final steps of translocation, which are required for factor dissociation and which are differentially regulated in bacterial and mammalian systems by the rates of deacyl-tRNA dissociation from the E site.


Subject(s)
Guanosine Triphosphate/metabolism , RNA, Transfer/metabolism , Ribosomes/metabolism , Animals , Bacteria/metabolism , Guanosine Triphosphate/chemistry , Humans , Hydrolysis , Internal Ribosome Entry Sites , Mammals/metabolism , Models, Molecular , Peptide Elongation Factor 2/chemistry , Peptide Elongation Factor 2/metabolism , Protein Domains , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Transfer/chemistry , Ribosomes/chemistry
7.
Open Biol ; 8(8)2018 08.
Article in English | MEDLINE | ID: mdl-30068566

ABSTRACT

Among cyclic nucleotide phosphodiesterases (PDEs), PDE6 is unique in serving as an effector enzyme in G protein-coupled signal transduction. In retinal rods and cones, PDE6 is membrane-bound and activated to hydrolyse its substrate, cGMP, by binding of two active G protein α-subunits (Gα*). To investigate the activation mechanism of mammalian rod PDE6, we have collected functional and structural data, and analysed them by reaction-diffusion simulations. Gα* titration of membrane-bound PDE6 reveals a strong functional asymmetry of the enzyme with respect to the affinity of Gα* for its two binding sites on membrane-bound PDE6 and the enzymatic activity of the intermediary 1 : 1 Gα* · PDE6 complex. Employing cGMP and its 8-bromo analogue as substrates, we find that Gα* · PDE6 forms with high affinity but has virtually no cGMP hydrolytic activity. To fully activate PDE6, it takes a second copy of Gα* which binds with lower affinity, forming Gα* · PDE6 · Gα*. Reaction-diffusion simulations show that the functional asymmetry of membrane-bound PDE6 constitutes a coincidence switch and explains the lack of G protein-related noise in visual signal transduction. The high local concentration of Gα* generated by a light-activated rhodopsin molecule efficiently activates PDE6, whereas the low density of spontaneously activated Gα* fails to activate the effector enzyme.


Subject(s)
Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Transducin/metabolism , Animals , Binding Sites , Cattle , Cell Membrane/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/chemistry , Enzyme Activation , Hydrolysis , Protein Binding , Transducin/chemistry
8.
Mol Cell ; 70(5): 881-893.e3, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29883607

ABSTRACT

The assembly of ribosomal subunits is an essential prerequisite for protein biosynthesis in all domains of life. Although biochemical and biophysical approaches have advanced our understanding of ribosome assembly, our mechanistic comprehension of this process is still limited. Here, we perform an in vitro reconstitution of the Escherichia coli 50S ribosomal subunit. Late reconstitution products were subjected to high-resolution cryo-electron microscopy and multiparticle refinement analysis to reconstruct five distinct precursors of the 50S subunit with 4.3-3.8 Å resolution. These assembly intermediates define a progressive maturation pathway culminating in a late assembly particle, whose structure is more than 96% identical to a mature 50S subunit. Our structures monitor the formation and stabilization of structural elements in a nascent particle in unprecedented detail and identify the maturation of the rRNA-based peptidyl transferase center as the final critical step along the 50S assembly pathway.


Subject(s)
Escherichia coli/metabolism , RNA, Bacterial/metabolism , RNA, Ribosomal, 23S/metabolism , Ribosome Subunits, Large, Bacterial/metabolism , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli/ultrastructure , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , RNA, Bacterial/genetics , RNA, Bacterial/ultrastructure , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 23S/ultrastructure , Ribosome Subunits, Large, Bacterial/genetics , Ribosome Subunits, Large, Bacterial/ultrastructure , Structure-Activity Relationship
9.
Nat Microbiol ; 2: 17062, 2017 Apr 28.
Article in English | MEDLINE | ID: mdl-28452979

ABSTRACT

λN-mediated processive antitermination constitutes a paradigmatic transcription regulatory event, during which phage protein λN, host factors NusA, NusB, NusE and NusG, and an RNA nut site render elongating RNA polymerase termination-resistant. The structural basis of the process has so far remained elusive. Here we describe a crystal structure of a λN-NusA-NusB-NusE-nut site complex and an electron cryo-microscopic structure of a complete transcription antitermination complex, comprising RNA polymerase, DNA, nut site RNA, all Nus factors and λN, validated by crosslinking/mass spectrometry. Due to intrinsic disorder, λN can act as a multiprotein/RNA interaction hub, which, together with nut site RNA, arranges NusA, NusB and NusE into a triangular complex. This complex docks via the NusA N-terminal domain and the λN C-terminus next to the RNA exit channel on RNA polymerase. Based on the structures, comparative crosslinking analyses and structure-guided mutagenesis, we hypothesize that λN mounts a multipronged strategy to reprogram the transcriptional machinery, which may include (1) the λN C terminus clamping the RNA exit channel, thus stabilizing the DNA:RNA hybrid; (2) repositioning of NusA and RNAP elements, thus redirecting nascent RNA and sequestering the upstream branch of a terminator hairpin; and (3) hindering RNA engagement of termination factor ρ and/or obstructing ρ translocation on the transcript.


Subject(s)
Bacterial Proteins/chemistry , DNA-Directed RNA Polymerases/chemistry , RNA-Binding Proteins/chemistry , Terminator Regions, Genetic , Transcription, Genetic , Binding Sites , DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Expression Regulation , RNA/chemistry , Rho Factor , Ribosomal Proteins/genetics , Transcription Factors/chemistry
10.
Nat Commun ; 7: 13521, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27995908

ABSTRACT

The surveillance of mRNA translation is imperative for homeostasis. Monitoring the integrity of the message is essential, as the translation of aberrant mRNAs leads to stalling of the translational machinery. During ribosomal rescue, arrested ribosomes are specifically recognized by the conserved eukaryotic proteins Dom34 and Hbs1, to initiate their recycling. Here we solve the structure of Dom34 and Hbs1 bound to a yeast ribosome programmed with a nonstop mRNA at 3.3 Å resolution using cryo-electron microscopy. The structure shows that Domain N of Dom34 is inserted into the upstream mRNA-binding groove via direct stacking interactions with conserved nucleotides of 18S rRNA. It senses the absence of mRNA at the A-site and part of the mRNA entry channel by direct competition. Thus, our analysis establishes the structural foundation for the recognition of aberrantly stalled 80S ribosomes by the Dom34·Hbs1·GTP complex during Dom34-mediated mRNA surveillance pathways.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Endoribonucleases/chemistry , Endoribonucleases/metabolism , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle Proteins/genetics , Cryoelectron Microscopy , Endoribonucleases/genetics , GTP-Binding Proteins/genetics , Guanylyl Imidodiphosphate/metabolism , HSP70 Heat-Shock Proteins/genetics , Models, Molecular , Peptide Elongation Factors/genetics , Protein Domains , Protein Interaction Domains and Motifs , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Static Electricity
11.
Sci Adv ; 2(3): e1501502, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26973877

ABSTRACT

Throughout the four phases of protein biosynthesis-initiation, elongation, termination, and recycling-the ribosome is controlled and regulated by at least one specified translational guanosine triphosphatase (trGTPase). Although the structural basis for trGTPase interaction with the ribosome has been solved for the last three steps of translation, the high-resolution structure for the key initiation trGTPase, initiation factor 2 (IF2), complexed with the ribosome, remains elusive. We determine the structure of IF2 complexed with a nonhydrolyzable guanosine triphosphate analog and initiator fMet-tRNAi (Met) in the context of the Escherichia coli ribosome to 3.7-Å resolution using cryo-electron microscopy. The structural analysis reveals previously unseen intrinsic conformational modes of the 70S initiation complex, establishing the mutual interplay of IF2 and initator transfer RNA (tRNA) with the ribsosome and providing the structural foundation for a mechanistic understanding of the final steps of translation initiation.


Subject(s)
Prokaryotic Initiation Factor-2/chemistry , Ribosomes/chemistry , Cryoelectron Microscopy , Models, Molecular , Protein Conformation
12.
EMBO J ; 34(24): 3042-58, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26604301

ABSTRACT

Internal ribosomal entry sites (IRESs) are structured cis-acting RNAs that drive an alternative, cap-independent translation initiation pathway. They are used by many viruses to hijack the translational machinery of the host cell. IRESs facilitate translation initiation by recruiting and actively manipulating the eukaryotic ribosome using only a subset of canonical initiation factor and IRES transacting factors. Here we present cryo-EM reconstructions of the ribosome 80S- and 40S-bound Hepatitis C Virus (HCV) IRES. The presence of four subpopulations for the 80S•HCV IRES complex reveals dynamic conformational modes of the complex. At a global resolution of 3.9 Šfor the most stable complex, a derived atomic model reveals a complex fold of the IRES RNA and molecular details of its interaction with the ribosome. The comparison of obtained structures explains how a modular architecture facilitates mRNA loading and tRNA binding to the P-site. This information provides the structural foundation for understanding the mechanism of HCV IRES RNA-driven translation initiation.


Subject(s)
Internal Ribosome Entry Sites , RNA, Viral/chemistry , Ribosome Subunits/chemistry , Amino Acid Sequence , Base Sequence , Hepatitis C/metabolism , Humans , Molecular Docking Simulation , Molecular Sequence Data , Protein Binding
13.
Methods Enzymol ; 558: 497-514, 2015.
Article in English | MEDLINE | ID: mdl-26068751

ABSTRACT

An explosion of new data from high-resolution cryo-electron microscopy (cryo-EM) studies has produced a large number of data sets for many species of ribosomes in various functional states over the past few years. While many methods exist to produce structural models for lower resolution cryo-EM reconstructions, high-resolution reconstructions are often modeled using crystallographic techniques and extensive manual intervention. Here, we present an automated fitting technique for high-resolution cryo-EM data sets that produces all-atom models highly consistent with the EM density. Using a molecular dynamics approach, atomic positions are optimized with a potential that includes the cross-correlation coefficient between the structural model and the cryo-EM electron density, as well as a biasing potential preserving the stereochemistry and secondary structure of the biomolecule. Specifically, we use a hybrid structure-based/ab initio molecular dynamics potential to extend molecular dynamics fitting. In addition, we find that simulated annealing integration, as opposed to straightforward molecular dynamics integration, significantly improves performance. We obtain atomistic models of the human ribosome consistent with high-resolution cryo-EM reconstructions of the human ribosome. Automated methods such as these have the potential to produce atomistic models for a large number of ribosome complexes simultaneously that can be subsequently refined manually.


Subject(s)
Molecular Dynamics Simulation/statistics & numerical data , RNA, Ribosomal/chemistry , Ribosomal Proteins/chemistry , Ribosome Subunits, Small, Eukaryotic/chemistry , Cryoelectron Microscopy , Humans , Kinetics , Nucleic Acid Conformation , Protein Conformation , Thermodynamics
14.
Cell ; 161(4): 845-57, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957688

ABSTRACT

Macromolecular machines, such as the ribosome, undergo large-scale conformational changes during their functional cycles. Although their mode of action is often compared to that of mechanical machines, a crucial difference is that, at the molecular dimension, thermodynamic effects dominate functional cycles, with proteins fluctuating stochastically between functional states defined by energetic minima on an energy landscape. Here, we have used cryo-electron microscopy to image ex-vivo-derived human polysomes as a source of actively translating ribosomes. Multiparticle refinement and 3D variability analysis allowed us to visualize a variety of native translation intermediates. Significantly populated states include not only elongation cycle intermediates in pre- and post-translocational states, but also eEF1A-containing decoding and termination/recycling complexes. Focusing on the post-translocational state, we extended this assessment to the single-residue level, uncovering striking details of ribosome-ligand interactions and identifying both static and functionally important dynamic elements.


Subject(s)
Protein Biosynthesis , Ribosomes/chemistry , Ribosomes/ultrastructure , Amino Acid Sequence , Cryoelectron Microscopy , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , RNA, Transfer/chemistry , Sequence Alignment , Thermodynamics
15.
Nat Struct Mol Biol ; 21(8): 721-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25064512

ABSTRACT

The universally conserved eukaryotic initiation factor (eIF) 5B, a translational GTPase, is essential for canonical translation initiation. It is also required for initiation facilitated by the internal ribosomal entry site (IRES) of hepatitis C virus (HCV) RNA. eIF5B promotes joining of 60S ribosomal subunits to 40S ribosomal subunits bound by initiator tRNA (Met-tRNAi(Met)). However, the exact molecular mechanism by which eIF5B acts has not been established. Here we present cryo-EM reconstructions of the mammalian 80S-HCV-IRES-Met-tRNAi(Met)-eIF5B-GMPPNP complex. We obtained two substates distinguished by the rotational state of the ribosomal subunits and the configuration of initiator tRNA in the peptidyl (P) site. Accordingly, a combination of conformational changes in the 80S ribosome and in initiator tRNA facilitates binding of the Met-tRNAi(Met) to the 60S P site and redefines the role of eIF5B as a tRNA-reorientation factor.


Subject(s)
Eukaryotic Initiation Factors/chemistry , Hepacivirus/genetics , RNA, Viral/chemistry , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/chemistry , Animals , Cryoelectron Microscopy , Fourier Analysis , Guanylyl Imidodiphosphate/chemistry , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , RNA, Transfer, Met/chemistry , Rabbits
16.
Cell ; 158(1): 121-31, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995983

ABSTRACT

The extent to which bacterial ribosomes and the significantly larger eukaryotic ribosomes share the same mechanisms of ribosomal elongation is unknown. Here, we present subnanometer resolution cryoelectron microscopy maps of the mammalian 80S ribosome in the posttranslocational state and in complex with the eukaryotic eEF1A⋅Val-tRNA⋅GMPPNP ternary complex, revealing significant differences in the elongation mechanism between bacteria and mammals. Surprisingly, and in contrast to bacterial ribosomes, a rotation of the small subunit around its long axis and orthogonal to the well-known intersubunit rotation distinguishes the posttranslocational state from the classical pretranslocational state ribosome. We term this motion "subunit rolling." Correspondingly, a mammalian decoding complex visualized in substates before and after codon recognition reveals structural distinctions from the bacterial system. These findings suggest how codon recognition leads to GTPase activation in the mammalian system and demonstrate that in mammalia subunit rolling occurs during tRNA selection.


Subject(s)
Mammals/metabolism , Ribosomes/chemistry , Amino Acid Sequence , Animals , Anticodon/metabolism , Codon/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Molecular Sequence Data , Peptide Chain Elongation, Translational , RNA, Transfer/metabolism , Rabbits , Saccharomyces cerevisiae/metabolism , Tetrahymena thermophila/metabolism
17.
Ultramicroscopy ; 140: 9-19, 2014 May.
Article in English | MEDLINE | ID: mdl-24562077

ABSTRACT

In structural electron microscopy, the accurate estimation of the Contrast Transfer Function (CTF) parameters, particularly defocus and astigmatism, is of utmost importance for both initial evaluation of micrograph quality and for subsequent structure determination. Due to increases in the rate of data collection on modern microscopes equipped with new generation cameras, it is also important that the CTF estimation can be done rapidly and with minimal user intervention. Finally, in order to minimize the necessity for manual screening of the micrographs by a user it is necessary to provide an assessment of the errors of fitted parameters values. In this work we introduce CTER, a CTF parameters estimation method distinguished by its computational efficiency. The efficiency of the method makes it suitable for high-throughput EM data collection, and enables the use of a statistical resampling technique, bootstrap, that yields standard deviations of estimated defocus and astigmatism amplitude and angle, thus facilitating the automation of the process of screening out inferior micrograph data. Furthermore, CTER also outputs the spatial frequency limit imposed by reciprocal space aliasing of the discrete form of the CTF and the finite window size. We demonstrate the efficiency and accuracy of CTER using a data set collected on a 300kV Tecnai Polara (FEI) using the K2 Summit DED camera in super-resolution counting mode. Using CTER we obtained a structure of the 80S ribosome whose large subunit had a resolution of 4.03Å without, and 3.85Å with, inclusion of astigmatism parameters.


Subject(s)
Cryoelectron Microscopy/methods , Image Processing, Computer-Assisted/methods , Algorithms , Cryoelectron Microscopy/statistics & numerical data , Image Processing, Computer-Assisted/statistics & numerical data , Imaging, Three-Dimensional/methods , Imaging, Three-Dimensional/statistics & numerical data , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Large, Eukaryotic/ultrastructure , Software
18.
Proc Natl Acad Sci U S A ; 110(52): 20964-9, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324168

ABSTRACT

During protein synthesis, coupled translocation of messenger RNAs (mRNA) and transfer RNAs (tRNA) through the ribosome takes place following formation of each peptide bond. The reaction is facilitated by large-scale conformational changes within the ribosomal complex and catalyzed by elongtion factor G (EF-G). Previous structural analysis of the interaction of EF-G with the ribosome used either model complexes containing no tRNA or only a single tRNA, or complexes where EF-G was directly bound to ribosomes in the posttranslocational state. Here, we present a multiparticle cryo-EM reconstruction of a translocation intermediate containing two tRNAs trapped in transit, bound in chimeric intrasubunit ap/P and pe/E hybrid states. The downstream ap/P-tRNA is contacted by domain IV of EF-G and P-site elements within the 30S subunit body, whereas the upstream pe/E-tRNA maintains tight interactions with P-site elements of the swiveled 30S head. Remarkably, a tight compaction of the tRNA pair can be seen in this state. The translocational intermediate presented here represents a previously missing link in understanding the mechanism of translocation, revealing that the ribosome uses two distinct molecular ratchets, involving both intra- and intersubunit rotational movements, to drive the synchronous movement of tRNAs and mRNA.


Subject(s)
Models, Molecular , Nucleic Acid Conformation , Peptide Chain Elongation, Translational/physiology , Peptide Elongation Factor G/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Escherichia coli , Image Processing, Computer-Assisted , RNA, Transfer/metabolism
19.
Nature ; 485(7399): 526-9, 2012 May 06.
Article in English | MEDLINE | ID: mdl-22622583

ABSTRACT

Bacterial ribosomes stalled at the 3' end of malfunctioning messenger RNAs can be rescued by transfer-messenger RNA (tmRNA)-mediated trans-translation. The SmpB protein forms a complex with the tmRNA, and the transfer-RNA-like domain (TLD) of the tmRNA then enters the A site of the ribosome. Subsequently, the TLD-SmpB module is translocated to the P site, a process that is facilitated by the elongation factor EF-G, and translation is switched to the mRNA-like domain (MLD) of the tmRNA. Accurate loading of the MLD into the mRNA path is an unusual initiation mechanism. Despite various snapshots of different ribosome-tmRNA complexes at low to intermediate resolution, it is unclear how the large, highly structured tmRNA is translocated and how the MLD is loaded. Here we present a cryo-electron microscopy reconstruction of a fusidic-acid-stalled ribosomal 70S-tmRNA-SmpB-EF-G complex (carrying both of the large ligands, that is, EF-G and tmRNA) at 8.3 Å resolution. This post-translocational intermediate (TI(POST)) presents the TLD-SmpB module in an intrasubunit ap/P hybrid site and a tRNA(fMet) in an intrasubunit pe/E hybrid site. Conformational changes in the ribosome and tmRNA occur in the intersubunit space and on the solvent side. The key underlying event is a unique extra-large swivel movement of the 30S head, which is crucial for both tmRNA-SmpB translocation and MLD loading, thereby coupling translocation to MLD loading. This mechanism exemplifies the versatile, dynamic nature of the ribosome, and it shows that the conformational modes of the ribosome that normally drive canonical translation can also be used in a modified form to facilitate more complex tasks in specialized non-canonical pathways.


Subject(s)
Escherichia coli/chemistry , Peptide Elongation Factor G/metabolism , Protein Biosynthesis , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA-Binding Proteins/metabolism , Ribosomes/metabolism , Base Sequence , Cryoelectron Microscopy , Fusidic Acid/metabolism , Ligands , Models, Molecular , Nucleic Acid Conformation , Peptide Elongation Factor G/chemistry , Peptide Elongation Factor G/ultrastructure , Protein Binding , Protein Conformation , RNA, Bacterial/genetics , RNA, Bacterial/ultrastructure , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/ultrastructure , Ribosome Subunits/chemistry , Ribosome Subunits/genetics , Ribosome Subunits/metabolism , Ribosome Subunits/ultrastructure , Ribosomes/chemistry , Ribosomes/genetics , Ribosomes/ultrastructure
20.
Nature ; 468(7324): 713-6, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21124459

ABSTRACT

The elongation cycle of protein synthesis involves the delivery of aminoacyl-transfer RNAs to the aminoacyl-tRNA-binding site (A site) of the ribosome, followed by peptide-bond formation and translocation of the tRNAs through the ribosome to reopen the A site. The translocation reaction is catalysed by elongation factor G (EF-G) in a GTP-dependent manner. Despite the availability of structures of various EF-G-ribosome complexes, the precise mechanism by which tRNAs move through the ribosome still remains unclear. Here we use multiparticle cryoelectron microscopy analysis to resolve two previously unseen subpopulations within Thermus thermophilus EF-G-ribosome complexes at subnanometre resolution, one of them with a partly translocated tRNA. Comparison of these substates reveals that translocation of tRNA on the 30S subunit parallels the swivelling of the 30S head and is coupled to unratcheting of the 30S body. Because the tRNA maintains contact with the peptidyl-tRNA-binding site (P site) on the 30S head and simultaneously establishes interaction with the exit site (E site) on the 30S platform, a novel intra-subunit 'pe/E' hybrid state is formed. This state is stabilized by domain IV of EF-G, which interacts with the swivelled 30S-head conformation. These findings provide direct structural and mechanistic insight into the 'missing link' in terms of tRNA intermediates involved in the universally conserved translocation process.


Subject(s)
Movement , RNA, Transfer/metabolism , Ribosome Subunits, Small, Bacterial/chemistry , Ribosome Subunits, Small, Bacterial/metabolism , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Models, Molecular , Peptide Elongation Factor G/chemistry , Peptide Elongation Factor G/metabolism , Protein Biosynthesis , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Transfer/chemistry , RNA, Transfer/ultrastructure , Ribosome Subunits, Small, Bacterial/ultrastructure , Thermus thermophilus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...