Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nat Struct Mol Biol ; 27(7): 625-634, 2020 07.
Article in English | MEDLINE | ID: mdl-32483338

ABSTRACT

Cyclic nucleotide-gated (CNG) channels convert cyclic nucleotide (CN) binding and unbinding into electrical signals in sensory receptors and neurons. The molecular conformational changes underpinning ligand activation are largely undefined. We report both closed- and open-state atomic cryo-EM structures of a full-length Caenorhabditis elegans cyclic GMP-activated channel TAX-4, reconstituted in lipid nanodiscs. These structures, together with computational and functional analyses and a mutant channel structure, reveal a double-barrier hydrophobic gate formed by two S6 amino acids in the central cavity. cGMP binding produces global conformational changes that open the cavity gate located ~52 Å away but do not alter the structure of the selectivity filter-the commonly presumed activation gate. Our work provides mechanistic insights into the allosteric gating and regulation of CN-gated and nucleotide-modulated channels and CNG channel-related channelopathies.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Ion Channels/chemistry , Ion Channels/metabolism , Caenorhabditis elegans Proteins/genetics , Cryoelectron Microscopy , Cyclic GMP/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Ion Channels/genetics , Ligands , Lipids/chemistry , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis , Mutation , Protein Conformation
2.
Sci Adv ; 6(14): eaay9572, 2020 04.
Article in English | MEDLINE | ID: mdl-32270040

ABSTRACT

The endoplasmic reticulum (ER) is a highly dynamic network of membranes. Here, we combine live-cell microscopy with in situ cryo-electron tomography to directly visualize ER dynamics in several secretory cell types including pancreatic ß-cells and neurons under near-native conditions. Using these imaging approaches, we identify a novel, mobile form of ER, ribosome-associated vesicles (RAVs), found primarily in the cell periphery, which is conserved across different cell types and species. We show that RAVs exist as distinct, highly dynamic structures separate from the intact ER reticular architecture that interact with mitochondria via direct intermembrane contacts. These findings describe a new ER subcompartment within cells.


Subject(s)
Cytoplasmic Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Ribosomes/metabolism , Animals , Biological Transport , Cryoelectron Microscopy , Cytoplasmic Vesicles/ultrastructure , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Molecular Imaging , Organ Specificity , Rats , Ribosomes/ultrastructure , Stress, Physiological
3.
Nat Commun ; 10(1): 2579, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189921

ABSTRACT

When the ribosome encounters a stop codon, it recruits a release factor (RF) to hydrolyze the ester bond between the peptide chain and tRNA. RFs have structural motifs that recognize stop codons in the decoding center and a GGQ motif for induction of hydrolysis in the peptidyl transfer center 70 Å away. Surprisingly, free RF2 is compact, with only 20 Å between its codon-reading and GGQ motifs. Cryo-EM showed that ribosome-bound RFs have extended structures, suggesting that RFs are compact when entering the ribosome and then extend their structures upon stop codon recognition. Here we use time-resolved cryo-EM to visualize transient compact forms of RF1 and RF2 at 3.5 and 4 Å resolution, respectively, in the codon-recognizing ribosome complex on the native pathway. About 25% of complexes have RFs in the compact state at 24 ms reaction time, and within 60 ms virtually all ribosome-bound RFs are transformed to their extended forms.


Subject(s)
Escherichia coli Proteins/ultrastructure , Models, Molecular , Peptide Chain Termination, Translational/physiology , Peptide Termination Factors/ultrastructure , Protein Domains/physiology , Binding Sites/physiology , Codon, Terminator/metabolism , Cryoelectron Microscopy , Escherichia coli Proteins/metabolism , Peptide Termination Factors/metabolism , RNA, Transfer/metabolism , Ribosome Subunits, Large, Bacterial/metabolism , Ribosome Subunits, Small, Bacterial/metabolism , Time Factors
4.
Proc Natl Acad Sci U S A ; 115(51): 12985-12990, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30509977

ABSTRACT

Membrane proteins function in native cell membranes, but extraction into isolated particles is needed for many biochemical and structural analyses. Commonly used detergent-extraction methods destroy naturally associated lipid bilayers. Here, we devised a detergent-free method for preparing cell-membrane nanoparticles to study the multidrug exporter AcrB, by cryo-EM at 3.2-Å resolution. We discovered a remarkably well-organized lipid-bilayer structure associated with transmembrane domains of the AcrB trimer. This bilayer patch comprises 24 lipid molecules; inner leaflet chains are packed in a hexagonal array, whereas the outer leaflet has highly irregular but ordered packing. Protein side chains interact with both leaflets and participate in the hexagonal pattern. We suggest that the lipid bilayer supports and harmonizes peristaltic motions through AcrB trimers. In AcrB D407A, a putative proton-relay mutant, lipid bilayer buttresses protein interactions lost in crystal structures after detergent-solubilization. Our detergent-free system preserves lipid-protein interactions for visualization and should be broadly applicable.


Subject(s)
Cell Membrane/metabolism , Detergents/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/metabolism , Cell Membrane/chemistry , Crystallography, X-Ray , Detergents/chemistry , Escherichia coli/growth & development , Nanoparticles/chemistry , Nanoparticles/metabolism , Protein Conformation
5.
Nature ; 553(7687): 233-237, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29258289

ABSTRACT

Calcium-selective transient receptor potential vanilloid subfamily member 6 (TRPV6) channels play a critical role in calcium uptake in epithelial tissues. Altered TRPV6 expression is associated with a variety of human diseases, including cancers. TRPV6 channels are constitutively active and their open probability depends on the lipidic composition of the membrane in which they reside; it increases substantially in the presence of phosphatidylinositol 4,5-bisphosphate. Crystal structures of detergent-solubilized rat TRPV6 in the closed state have previously been solved. Corroborating electrophysiological results, these structures demonstrated that the Ca2+ selectivity of TRPV6 arises from a ring of aspartate side chains in the selectivity filter that binds Ca2+ tightly. However, how TRPV6 channels open and close their pores for ion permeation has remained unclear. Here we present cryo-electron microscopy structures of human TRPV6 in the open and closed states. The channel selectivity filter adopts similar conformations in both states, consistent with its explicit role in ion permeation. The iris-like channel opening is accompanied by an α-to-π-helical transition in the pore-lining transmembrane helix S6 at an alanine hinge just below the selectivity filter. As a result of this transition, the S6 helices bend and rotate, exposing different residues to the ion channel pore in the open and closed states. This gating mechanism, which defines the constitutive activity of TRPV6, is, to our knowledge, unique among tetrameric ion channels and provides structural insights for understanding their diverse roles in physiology and disease.


Subject(s)
Calcium Channels/metabolism , Calcium Channels/ultrastructure , Cryoelectron Microscopy , Epithelial Cells/metabolism , Ion Channel Gating , TRPV Cation Channels/metabolism , TRPV Cation Channels/ultrastructure , Alanine/metabolism , Calcium/metabolism , Calcium Channels/chemistry , Humans , Ion Transport , Protein Conformation , Rotation , TRPV Cation Channels/chemistry
6.
Nature ; 549(7670): 60-65, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28737760

ABSTRACT

AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)-subtype ionotropic glutamate receptors mediate fast excitatory neurotransmission throughout the central nervous system. Gated by the neurotransmitter glutamate, AMPA receptors are critical for synaptic strength, and dysregulation of AMPA receptor-mediated signalling is linked to numerous neurological diseases. Here we use cryo-electron microscopy to solve the structures of AMPA receptor-auxiliary subunit complexes in the apo, antagonist- and agonist-bound states and determine the iris-like mechanism of ion channel opening. The ion channel selectivity filter is formed by the extended portions of the re-entrant M2 loops, while the helical portions of M2 contribute to extensive hydrophobic interfaces between AMPA receptor subunits in the ion channel. We show how the permeation pathway changes upon channel opening and identify conformational changes throughout the entire AMPA receptor that accompany activation and desensitization. Our findings provide a framework for understanding gating across the family of ionotropic glutamate receptors and the role of AMPA receptors in excitatory neurotransmission.


Subject(s)
Cryoelectron Microscopy , Ion Channel Gating , Receptors, AMPA/chemistry , Receptors, AMPA/ultrastructure , Animals , Calcium Channels/metabolism , Claudins/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Protein Conformation , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Synaptic Transmission
7.
Neuron ; 94(3): 569-580.e5, 2017 May 03.
Article in English | MEDLINE | ID: mdl-28472657

ABSTRACT

Fast excitatory neurotransmission is mediated by AMPA-subtype ionotropic glutamate receptors (AMPARs). AMPARs, localized at post-synaptic densities, are regulated by transmembrane auxiliary subunits that modulate AMPAR assembly, trafficking, gating, and pharmacology. Aberrancies in AMPAR-mediated signaling are associated with numerous neurological disorders. Here, we report cryo-EM structures of an AMPAR in complex with the auxiliary subunit GSG1L in the closed and desensitized states. GSG1L favors the AMPAR desensitized state, where channel closure is facilitated by profound structural rearrangements in the AMPAR extracellular domain, with ligand-binding domain dimers losing their local 2-fold rotational symmetry. Our structural and functional experiments suggest that AMPAR auxiliary subunits share a modular architecture and use a common transmembrane scaffold for distinct extracellular modules to differentially regulate AMPAR gating. By comparing the AMPAR-GSG1L complex structures, we map conformational changes accompanying AMPAR recovery from desensitization and reveal structural bases for regulation of synaptic transmission by auxiliary subunits.


Subject(s)
Claudins/metabolism , Protein Structure, Quaternary , Protein Subunits/metabolism , Receptors, AMPA/metabolism , Animals , Calcium Channels/metabolism , Cryoelectron Microscopy , HEK293 Cells , Humans , Ion Channel Gating , Mice , Models, Molecular , Post-Synaptic Density/metabolism , Protein Binding , Protein Transport , Rats , Sf9 Cells , Spodoptera
8.
Structure ; 25(4): 663-670.e3, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28286002

ABSTRACT

We describe a spraying-plunging method for preparing cryoelectron microscopy (cryo-EM) grids with vitreous ice of controllable, highly consistent thickness using a microfluidic device. The new polydimethylsiloxane (PDMS)-based sprayer was tested with apoferritin. We demonstrate that the structure can be solved to high resolution with this method of sample preparation. Besides replacing the conventional pipetting-blotting-plunging method, one of many potential applications of the new sprayer is in time-resolved cryo-EM, as part of a PDMS-based microfluidic reaction channel to study short-lived intermediates on the timescale of 10-1,000 ms.


Subject(s)
Cryoelectron Microscopy/methods , Microfluidics/instrumentation , Dimethylpolysiloxanes/chemistry , Microfluidics/methods
9.
Protein Sci ; 26(1): 82-92, 2017 01.
Article in English | MEDLINE | ID: mdl-27750394

ABSTRACT

With the advance of new instruments and algorithms, and the accumulation of experience over decades, single-particle cryo-EM has become a pivotal part of structural biology. Recently, we determined the structure of a eukaryotic ribosome at 2.5 Å for the large subunit. The ribosome was derived from Trypanosoma cruzi, the protozoan pathogen of Chagas disease. The high-resolution density map allowed us to discern a large number of unprecedented details including rRNA modifications, water molecules, and ions such as Mg2+ and Zn2+ . In this paper, we focus on the procedures for data collection, image processing, and modeling, with particular emphasis on factors that contributed to the attainment of high resolution. The methods described here are readily applicable to other macromolecules for high-resolution reconstruction by single-particle cryo-EM.


Subject(s)
Cryoelectron Microscopy/methods , RNA Processing, Post-Transcriptional , RNA, Protozoan/ultrastructure , RNA, Ribosomal/ultrastructure , Ribosomes/ultrastructure , Trypanosoma cruzi/ultrastructure , Chagas Disease , Humans , Magnesium/metabolism , RNA, Protozoan/metabolism , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Trypanosoma cruzi/metabolism , Zinc/metabolism
10.
Structure ; 24(12): 2092-2101, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27818103

ABSTRACT

Upon encountering a stop codon on mRNA, polypeptide synthesis on the ribosome is terminated by release factors, and the ribosome complex, still bound with mRNA and P-site-bound tRNA (post-termination complex, PostTC), is split into ribosomal subunits, ready for a new round of translational initiation. Separation of post-termination ribosomes into subunits, or "ribosome recycling," is promoted by the joint action of ribosome-recycling factor (RRF) and elongation factor G (EF-G) in a guanosine triphosphate (GTP) hydrolysis-dependent manner. Here we used a mixing-spraying-based method of time-resolved cryo-electron microscopy (cryo-EM) to visualize the short-lived intermediates of the recycling process. The two complexes that contain (1) both RRF and EF-G bound to the PostTC or (2) deacylated tRNA bound to the 30S subunit are of particular interest. Our observations of the native form of these complexes demonstrate the strong potential of time-resolved cryo-EM for visualizing previously unobservable transient structures.


Subject(s)
Escherichia coli/metabolism , Peptide Elongation Factor G/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Binding Sites , Cryoelectron Microscopy , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Peptide Elongation Factor G/chemistry , Protein Binding , Protein Biosynthesis , RNA, Transfer/metabolism , Ribosomal Proteins/chemistry
11.
Proc Natl Acad Sci U S A ; 113(43): 12174-12179, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27791004

ABSTRACT

Ribosomes of trypanosomatids, a family of protozoan parasites causing debilitating human diseases, possess multiply fragmented rRNAs that together are analogous to 28S rRNA, unusually large rRNA expansion segments, and r-protein variations compared with other eukaryotic ribosomes. To investigate the architecture of the trypanosomatid ribosomes, we determined the 2.5-Å structure of the Trypanosoma cruzi ribosome large subunit by single-particle cryo-EM. Examination of this structure and comparative analysis of the yeast ribosomal assembly pathway allowed us to develop a stepwise assembly model for the eight pieces of the large subunit rRNAs and a number of ancillary "glue" proteins. This model can be applied to the characterization of Trypanosoma brucei and Leishmania spp. ribosomes as well. Together with other details, our atomic-level structure may provide a foundation for structure-based design of antitrypanosome drugs.


Subject(s)
Ribosome Subunits, Large, Eukaryotic/ultrastructure , Ribosomes/ultrastructure , Trypanosoma cruzi/chemistry , Chagas Disease/drug therapy , Chagas Disease/parasitology , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , RNA, Ribosomal/genetics , RNA, Ribosomal/ultrastructure , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Large, Eukaryotic/genetics , Ribosomes/genetics , Trypanosoma cruzi/genetics , Trypanosoma cruzi/ultrastructure
12.
Cell ; 167(1): 145-157.e17, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662087

ABSTRACT

The type-1 ryanodine receptor (RyR1) is an intracellular calcium (Ca(2+)) release channel required for skeletal muscle contraction. Here, we present cryo-EM reconstructions of RyR1 in multiple functional states revealing the structural basis of channel gating and ligand-dependent activation. Binding sites for the channel activators Ca(2+), ATP, and caffeine were identified at interdomain interfaces of the C-terminal domain. Either ATP or Ca(2+) alone induces conformational changes in the cytoplasmic assembly ("priming"), without pore dilation. In contrast, in the presence of all three activating ligands, high-resolution reconstructions of open and closed states of RyR1 were obtained from the same sample, enabling analyses of conformational changes associated with gating. Gating involves global conformational changes in the cytosolic assembly accompanied by local changes in the transmembrane domain, which include bending of the S6 transmembrane segment and consequent pore dilation, displacement, and deformation of the S4-S5 linker and conformational changes in the pseudo-voltage-sensor domain.


Subject(s)
Calcium Channel Agonists/chemistry , Ion Channel Gating , Muscle Contraction , Ryanodine Receptor Calcium Release Channel/chemistry , Animals , Binding Sites , Caffeine/chemistry , Calcium/chemistry , Cryoelectron Microscopy , Ligands , Protein Domains , Rabbits , Tacrolimus Binding Proteins/chemistry
13.
Science ; 353(6294): 83-6, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27365450

ABSTRACT

AMPA-subtype ionotropic glutamate receptors (AMPARs) mediate fast excitatory neurotransmission and contribute to high cognitive processes such as learning and memory. In the brain, AMPAR trafficking, gating, and pharmacology is tightly controlled by transmembrane AMPAR regulatory proteins (TARPs). Here, we used cryo-electron microscopy to elucidate the structural basis of AMPAR regulation by one of these auxiliary proteins, TARP γ2, or stargazin (STZ). Our structures illuminate the variable interaction stoichiometry of the AMPAR-TARP complex, with one or two TARP molecules binding one tetrameric AMPAR. Analysis of the AMPAR-STZ binding interfaces suggests that electrostatic interactions between the extracellular domains of AMPAR and STZ play an important role in modulating AMPAR function through contact surfaces that are conserved across AMPARs and TARPs. We propose a model explaining how TARPs stabilize the activated state of AMPARs and how the interactions between AMPARs and their auxiliary proteins control fast excitatory synaptic transmission.


Subject(s)
Brain/metabolism , Calcium Channels/chemistry , Receptors, AMPA/chemistry , Synaptic Transmission , Animals , Calcium Channels/ultrastructure , Cryoelectron Microscopy , HEK293 Cells , Humans , Models, Molecular , Protein Stability , Protein Structure, Secondary , Rats , Receptors, AMPA/ultrastructure
14.
J Struct Biol ; 190(3): 348-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25913484

ABSTRACT

Image formation in bright field electron microscopy can be described with the help of the contrast transfer function (CTF). In this work the authors describe the "CTF Estimation Challenge", called by the Madrid Instruct Image Processing Center (I2PC) in collaboration with the National Center for Macromolecular Imaging (NCMI) at Houston. Correcting for the effects of the CTF requires accurate knowledge of the CTF parameters, but these have often been difficult to determine. In this challenge, researchers have had the opportunity to test their ability in estimating some of the key parameters of the electron microscope CTF on a large micrograph data set produced by well-known laboratories on a wide set of experimental conditions. This work presents the first analysis of the results of the CTF Estimation Challenge, including an assessment of the performance of the different software packages under different conditions, so as to identify those areas of research where further developments would be desirable in order to achieve high-resolution structural information.


Subject(s)
Macromolecular Substances/chemistry , Microscopy, Electron/methods , Algorithms , Image Processing, Computer-Assisted/methods , Software
15.
Nature ; 517(7532): 44-9, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25470061

ABSTRACT

Ryanodine receptors (RyRs) mediate the rapid release of calcium (Ca(2+)) from intracellular stores into the cytosol, which is essential for numerous cellular functions including excitation-contraction coupling in muscle. Lack of sufficient structural detail has impeded understanding of RyR gating and regulation. Here we report the closed-state structure of the 2.3-megadalton complex of the rabbit skeletal muscle type 1 RyR (RyR1), solved by single-particle electron cryomicroscopy at an overall resolution of 4.8 Å. We fitted a polyalanine-level model to all 3,757 ordered residues in each protomer, defining the transmembrane pore in unprecedented detail and placing all cytosolic domains as tertiary folds. The cytosolic assembly is built on an extended α-solenoid scaffold connecting key regulatory domains to the pore. The RyR1 pore architecture places it in the six-transmembrane ion channel superfamily. A unique domain inserted between the second and third transmembrane helices interacts intimately with paired EF-hands originating from the α-solenoid scaffold, suggesting a mechanism for channel gating by Ca(2+).


Subject(s)
Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/ultrastructure , Animals , Calcium/deficiency , Calcium/metabolism , Calcium/pharmacology , Cell Membrane/metabolism , Cryoelectron Microscopy , Cytosol/metabolism , Ion Channel Gating/drug effects , Muscle, Skeletal/chemistry , Protein Structure, Tertiary , Rabbits , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/metabolism
16.
Nucleic Acids Res ; 42(5): 3409-18, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24335085

ABSTRACT

Eukaryotic translation termination results from the complex functional interplay between two release factors, eRF1 and eRF3, in which GTP hydrolysis by eRF3 couples codon recognition with peptidyl-tRNA hydrolysis by eRF1. Here, we present a cryo-electron microscopy structure of pre-termination complexes associated with eRF1•eRF3•GDPNP at 9.7 -Šresolution, which corresponds to the initial pre-GTP hydrolysis stage of factor attachment and stop codon recognition. It reveals the ribosomal positions of eRFs and provides insights into the mechanisms of stop codon recognition and triggering of eRF3's GTPase activity.


Subject(s)
Peptide Chain Termination, Translational , Peptide Termination Factors/chemistry , Ribosomes/chemistry , Codon, Terminator , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/chemistry , Humans , Models, Molecular , Peptide Termination Factors/metabolism
17.
Nature ; 503(7477): 539-43, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24185006

ABSTRACT

Hepatitis C virus (HCV) and classical swine fever virus (CSFV) messenger RNAs contain related (HCV-like) internal ribosome entry sites (IRESs) that promote 5'-end independent initiation of translation, requiring only a subset of the eukaryotic initiation factors (eIFs) needed for canonical initiation on cellular mRNAs. Initiation on HCV-like IRESs relies on their specific interaction with the 40S subunit, which places the initiation codon into the P site, where it directly base-pairs with eIF2-bound initiator methionyl transfer RNA to form a 48S initiation complex. However, all HCV-like IRESs also specifically interact with eIF3 (refs 2, 5-7, 9-12), but the role of this interaction in IRES-mediated initiation has remained unknown. During canonical initiation, eIF3 binds to the 40S subunit as a component of the 43S pre-initiation complex, and comparison of the ribosomal positions of eIF3 and the HCV IRES revealed that they overlap, so that their rearrangement would be required for formation of ribosomal complexes containing both components. Here we present a cryo-electron microscopy reconstruction of a 40S ribosomal complex containing eIF3 and the CSFV IRES. Remarkably, although the position and interactions of the CSFV IRES with the 40S subunit in this complex are similar to those of the HCV IRES in the 40S-IRES binary complex, eIF3 is completely displaced from its ribosomal position in the 43S complex, and instead interacts through its ribosome-binding surface exclusively with the apical region of domain III of the IRES. Our results suggest a role for the specific interaction of HCV-like IRESs with eIF3 in preventing ribosomal association of eIF3, which could serve two purposes: relieving the competition between the IRES and eIF3 for a common binding site on the 40S subunit, and reducing formation of 43S complexes, thereby favouring translation of viral mRNAs.


Subject(s)
Classical Swine Fever Virus/genetics , Eukaryotic Initiation Factor-3/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Regulatory Sequences, Ribonucleic Acid/genetics , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosomes/metabolism , Animals , Binding, Competitive , Cryoelectron Microscopy , Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-3/ultrastructure , Humans , Models, Molecular , Protein Biosynthesis , Rabbits , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/ultrastructure , Ribosomes/chemistry , Ribosomes/ultrastructure
18.
Cell ; 153(5): 1108-19, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706745

ABSTRACT

Eukaryotic translation initiation begins with assembly of a 43S preinitiation complex. First, methionylated initiator methionine transfer RNA (Met-tRNAi(Met)), eukaryotic initiation factor (eIF) 2, and guanosine triphosphate form a ternary complex (TC). The TC, eIF3, eIF1, and eIF1A cooperatively bind to the 40S subunit, yielding the 43S preinitiation complex, which is ready to attach to messenger RNA (mRNA) and start scanning to the initiation codon. Scanning on structured mRNAs additionally requires DHX29, a DExH-box protein that also binds directly to the 40S subunit. Here, we present a cryo-electron microscopy structure of the mammalian DHX29-bound 43S complex at 11.6 Å resolution. It reveals that eIF2 interacts with the 40S subunit via its α subunit and supports Met-tRNAi(Met) in an unexpected P/I orientation (eP/I). The structural core of eIF3 resides on the back of the 40S subunit, establishing two principal points of contact, whereas DHX29 binds around helix 16. The structure provides insights into eukaryote-specific aspects of translation, including the mechanism of action of DHX29.


Subject(s)
Mammals/metabolism , Peptide Chain Initiation, Translational , RNA Helicases/chemistry , RNA, Ribosomal/chemistry , Ribonucleoproteins/chemistry , Animals , Base Sequence , Cell-Free System , Cryoelectron Microscopy , Eukaryotic Initiation Factor-2/chemistry , Eukaryotic Initiation Factor-2/metabolism , Humans , Mammals/genetics , Models, Molecular , Molecular Sequence Data , RNA Helicases/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal, 18S/chemistry , RNA, Ribosomal, 18S/metabolism , Rabbits , Ribonucleoproteins/metabolism
19.
Nature ; 494(7437): 385-9, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23395961

ABSTRACT

Ribosomes, the protein factories of living cells, translate genetic information carried by messenger RNAs into proteins, and are thus involved in virtually all aspects of cellular development and maintenance. The few available structures of the eukaryotic ribosome reveal that it is more complex than its prokaryotic counterpart, owing mainly to the presence of eukaryote-specific ribosomal proteins and additional ribosomal RNA insertions, called expansion segments. The structures also differ among species, partly in the size and arrangement of these expansion segments. Such differences are extreme in kinetoplastids, unicellular eukaryotic parasites often infectious to humans. Here we present a high-resolution cryo-electron microscopy structure of the ribosome of Trypanosoma brucei, the parasite that is transmitted by the tsetse fly and that causes African sleeping sickness. The atomic model reveals the unique features of this ribosome, characterized mainly by the presence of unusually large expansion segments and ribosomal-protein extensions leading to the formation of four additional inter-subunit bridges. We also find additional rRNA insertions, including one large rRNA domain that is not found in other eukaryotes. Furthermore, the structure reveals the five cleavage sites of the kinetoplastid large ribosomal subunit (LSU) rRNA chain, which is known to be cleaved uniquely into six pieces, and suggests that the cleavage is important for the maintenance of the T. brucei ribosome in the observed structure. We discuss several possible implications of the large rRNA expansion segments for the translation-regulation process. The structure could serve as a basis for future experiments aimed at understanding the functional importance of these kinetoplastid-specific ribosomal features in protein-translation regulation, an essential step towards finding effective and safe kinetoplastid-specific drugs.


Subject(s)
Cryoelectron Microscopy , Ribosomes/ultrastructure , Trypanosoma brucei brucei/cytology , Trypanosoma brucei brucei/ultrastructure , Models, Biological , Models, Molecular , Molecular Conformation , Protein Biosynthesis , RNA, Protozoan/genetics , RNA, Protozoan/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Ribosomes/chemistry , Ribosomes/genetics , Trypanosoma brucei brucei/chemistry , Trypanosoma brucei brucei/genetics , Yeasts/chemistry
20.
Proc Natl Acad Sci U S A ; 109(45): 18413-8, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23091004

ABSTRACT

Eukaryotic translation termination results from the complex functional interplay between two eukaryotic release factors, eRF1 and eRF3, and the ribosome, in which GTP hydrolysis by eRF3 couples codon recognition with peptidyl-tRNA hydrolysis by eRF1. Here, using cryo-electron microscopy (cryo-EM) and flexible fitting, we determined the structure of eRF1-eRF3-guanosine 5'-[ß,γ-imido]triphosphate (GMPPNP)-bound ribosomal pretermination complex (pre-TC), which corresponds to the initial, pre-GTP hydrolysis stage of factor attachment. Our results show that eukaryotic translation termination involves a network of interactions between the two release factors and the ribosome. Our structure provides mechanistic insight into the coordination between GTP hydrolysis by eRF3 and subsequent peptide release by eRF1.


Subject(s)
Cryoelectron Microscopy , Mammals/metabolism , Peptide Chain Termination, Translational , Peptide Termination Factors/ultrastructure , Animals , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Models, Molecular , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Protein Binding , Protein Conformation , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , Rabbits , Ribosomes/metabolism , Ribosomes/ultrastructure , Saccharomyces cerevisiae
SELECTION OF CITATIONS
SEARCH DETAIL
...