Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
Add more filters

Publication year range
1.
Cell ; 187(3): 782-796.e23, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38244547

ABSTRACT

The rapid kinetics of biological processes and associated short-lived conformational changes pose a significant challenge in attempts to structurally visualize biomolecules during a reaction in real time. Conventionally, on-pathway intermediates have been trapped using chemical modifications or reduced temperature, giving limited insights. Here, we introduce a time-resolved cryo-EM method using a reusable PDMS-based microfluidic chip assembly with high reactant mixing efficiency. Coating of PDMS walls with SiO2 virtually eliminates non-specific sample adsorption and ensures maintenance of the stoichiometry of the reaction, rendering it highly reproducible. In an operating range from 10 to 1,000 ms, the device allows us to follow in vitro reactions of biological molecules at resolution levels in the range of 3 Å. By employing this method, we show the mechanism of progressive HflX-mediated splitting of the 70S E. coli ribosome in the presence of the GTP via capture of three high-resolution reaction intermediates within 140 ms.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Ribosomes , Cryoelectron Microscopy/methods , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , GTP-Binding Proteins/metabolism , Microfluidics/methods , Ribosomes/metabolism , Silicon Dioxide/analysis
2.
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
3.
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
4.
Trends Biochem Sci ; 47(2): 117-123, 2022 02.
Article in English | MEDLINE | ID: mdl-34799235

ABSTRACT

Single-particle cryoelectron microscopy (cryo-EM), whose full capabilities have been realized only within the past decade, has had a pivotal role in the fight against COVID-19. This is due to the technique's intrinsic power to depict both structural and dynamic features of molecules; in this case, of the spike protein of SARS-CoV-2. By now, numerous cryo-EM studies have furthered our understanding of spike protein-angiotensin-converting enzyme 2 (ACE2) receptor interactions, which has informed the design of effective vaccines, and have enabled the characterization of neutralizing antibody binding sites, which will lead to the design of novel therapeutics as the virus evolves.


Subject(s)
COVID-19 , Spike Glycoprotein, Coronavirus , Antibodies, Neutralizing/metabolism , Cryoelectron Microscopy , Humans , Protein Binding , SARS-CoV-2
5.
EMBO J ; 41(16): e110581, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35822879

ABSTRACT

Hepatitis C virus mRNA contains an internal ribosome entry site (IRES) that mediates end-independent translation initiation, requiring a subset of eukaryotic initiation factors (eIFs). Biochemical studies revealed that direct binding of the IRES to the 40S ribosomal subunit places the initiation codon into the P site, where it base pairs with eIF2-bound Met-tRNAiMet forming a 48S initiation complex. Subsequently, eIF5 and eIF5B mediate subunit joining, yielding an elongation-competent 80S ribosome. Initiation can also proceed without eIF2, in which case Met-tRNAiMet is recruited directly by eIF5B. However, the structures of initiation complexes assembled on the HCV IRES, the transitions between different states, and the accompanying conformational changes have remained unknown. To fill these gaps, we now obtained cryo-EM structures of IRES initiation complexes, at resolutions up to 3.5 Å, that cover all major stages from the initial ribosomal association, through eIF2-containing 48S initiation complexes, to eIF5B-containing complexes immediately prior to subunit joining. These structures provide insights into the dynamic network of 40S/IRES contacts, highlight the role of IRES domain II, and reveal conformational changes that occur during the transition from eIF2- to eIF5B-containing 48S complexes and prepare them for subunit joining.


Subject(s)
Hepacivirus , Hepatitis C , Eukaryotic Initiation Factor-2/metabolism , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/metabolism , Humans , Internal Ribosome Entry Sites , Protein Biosynthesis , RNA, Viral/genetics , RNA, Viral/metabolism , Ribosomes/metabolism
7.
Nature ; 584(7820): 304-309, 2020 08.
Article in English | MEDLINE | ID: mdl-32581365

ABSTRACT

The human GABAB receptor-a member of the class C family of G-protein-coupled receptors (GPCRs)-mediates inhibitory neurotransmission and has been implicated in epilepsy, pain and addiction1. A unique GPCR that is known to require heterodimerization for function2-6, the GABAB receptor has two subunits, GABAB1 and GABAB2, that are structurally homologous but perform distinct and complementary functions. GABAB1 recognizes orthosteric ligands7,8, while GABAB2 couples with G proteins9-14. Each subunit is characterized by an extracellular Venus flytrap (VFT) module, a descending peptide linker, a seven-helix transmembrane domain and a cytoplasmic tail15. Although the VFT heterodimer structure has been resolved16, the structure of the full-length receptor and its transmembrane signalling mechanism remain unknown. Here we present a near full-length structure of the GABAB receptor, captured in an inactive state by cryo-electron microscopy. Our structure reveals several ligands that preassociate with the receptor, including two large endogenous phospholipids that are embedded within the transmembrane domains to maintain receptor integrity and modulate receptor function. We also identify a previously unknown heterodimer interface between transmembrane helices 3 and 5 of both subunits, which serves as a signature of the inactive conformation. A unique 'intersubunit latch' within this transmembrane interface maintains the inactive state, and its disruption leads to constitutive receptor activity.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-B/chemistry , Receptors, GABA-B/ultrastructure , Calcium/metabolism , Ethanolamines/chemistry , Ethanolamines/metabolism , Humans , Ligands , Models, Molecular , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Protein Domains , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, GABA-B/metabolism , Structure-Activity Relationship
8.
Annu Rev Biochem ; 79: 381-412, 2010.
Article in English | MEDLINE | ID: mdl-20235828

ABSTRACT

There is mounting evidence indicating that protein synthesis is driven and regulated by mechanisms that direct stochastic, large-scale conformational fluctuations of the translational apparatus. This mechanistic paradigm implies that a free-energy landscape governs the conformational states that are accessible to and sampled by the translating ribosome. This scenario presents interdependent opportunities and challenges for structural and dynamic studies of protein synthesis. Indeed, the synergism between cryogenic electron microscopic and X-ray crystallographic structural studies, on the one hand, and single-molecule fluorescence resonance energy transfer (smFRET) dynamic studies, on the other, is emerging as a powerful means for investigating the complex free-energy landscape of the translating ribosome and uncovering the mechanisms that direct the stochastic conformational fluctuations of the translational machinery. In this review, we highlight the principal insights obtained from cryogenic electron microscopic, X-ray crystallographic, and smFRET studies of the elongation stage of protein synthesis and outline the emerging themes, questions, and challenges that lie ahead in mechanistic studies of translation.


Subject(s)
Peptide Chain Elongation, Translational , Ribosomes/chemistry , Ribosomes/metabolism , Animals , Cryoelectron Microscopy , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Humans , Protein Biosynthesis , RNA, Transfer/metabolism
9.
Nature ; 570(7761): 400-404, 2019 06.
Article in English | MEDLINE | ID: mdl-31108498

ABSTRACT

The initiation of bacterial translation involves the tightly regulated joining of the 50S ribosomal subunit to an initiator transfer RNA (fMet-tRNAfMet)-containing 30S ribosomal initiation complex to form a 70S initiation complex, which subsequently matures into a 70S elongation-competent complex. Rapid and accurate formation of the 70S initiation complex is promoted by initiation factors, which must dissociate from the 30S initiation complex before the resulting 70S elongation-competent complex can begin the elongation of translation1. Although comparisons of the structures of the 30S2-5 and 70S4,6-8 initiation complexes have revealed that the ribosome, initiation factors and fMet-tRNAfMet can acquire different conformations in these complexes, the timing of conformational changes during formation of the 70S initiation complex, the structures of any intermediates formed during these rearrangements, and the contributions that these dynamics might make to the mechanism and regulation of initiation remain unknown. Moreover, the absence of a structure of the 70S elongation-competent complex formed via an initiation-factor-catalysed reaction has precluded an understanding of the rearrangements to the ribosome, initiation factors and fMet-tRNAfMet that occur during maturation of a 70S initiation complex into a 70S elongation-competent complex. Here, using time-resolved cryogenic electron microscopy9, we report the near-atomic-resolution view of how a time-ordered series of conformational changes drive and regulate subunit joining, initiation factor dissociation and fMet-tRNAfMet positioning during formation of the 70S elongation-competent complex. Our results demonstrate the power of time-resolved cryogenic electron microscopy to determine how a time-ordered series of conformational changes contribute to the mechanism and regulation of one of the most fundamental processes in biology.


Subject(s)
Cryoelectron Microscopy , Escherichia coli/metabolism , Escherichia coli/ultrastructure , Peptide Chain Initiation, Translational , Ribosomes/metabolism , Ribosomes/ultrastructure , Escherichia coli/chemistry , Peptide Chain Elongation, Translational , Protein Conformation , Ribosome Subunits, Large, Bacterial/metabolism , Ribosome Subunits, Large, Bacterial/ultrastructure , Ribosome Subunits, Small, Bacterial/metabolism , Ribosome Subunits, Small, Bacterial/ultrastructure , Ribosomes/chemistry , Time Factors
10.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34916296

ABSTRACT

The human extracellular calcium-sensing (CaS) receptor controls plasma Ca2+ levels and contributes to nutrient-dependent maintenance and metabolism of diverse organs. Allosteric modulation of the CaS receptor corrects disorders of calcium homeostasis. Here, we report the cryogenic-electron microscopy reconstructions of a near-full-length CaS receptor in the absence and presence of allosteric modulators. Activation of the homodimeric CaS receptor requires a break in the transmembrane 6 (TM6) helix of each subunit, which facilitates the formation of a TM6-mediated homodimer interface and expansion of homodimer interactions. This transformation in TM6 occurs without a positive allosteric modulator. Two modulators with opposite functional roles bind to overlapping sites within the transmembrane domain through common interactions, acting to stabilize distinct rotamer conformations of key residues on the TM6 helix. The positive modulator reinforces TM6 distortion and maximizes subunit contact to enhance receptor activity, while the negative modulator strengthens an intact TM6 to dampen receptor function. In both active and inactive states, the receptor displays symmetrical transmembrane conformations that are consistent with its homodimeric assembly.


Subject(s)
Calcium/metabolism , Gene Expression Regulation/physiology , Homeostasis/physiology , Receptors, Calcium-Sensing/metabolism , Cryoelectron Microscopy , HEK293 Cells , Humans , Models, Molecular , Protein Conformation , Protein Domains , Receptors, Calcium-Sensing/genetics , Signal Transduction
11.
EMBO J ; 38(21): e102226, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31609474

ABSTRACT

Colony collapse disorder (CCD) is a multi-faceted syndrome decimating bee populations worldwide, and a group of viruses of the widely distributed Dicistroviridae family have been identified as a causing agent of CCD. This family of viruses employs non-coding RNA sequences, called internal ribosomal entry sites (IRESs), to precisely exploit the host machinery for viral protein production. Using single-particle cryo-electron microscopy (cryo-EM), we have characterized how the IRES of Israeli acute paralysis virus (IAPV) intergenic region captures and redirects translating ribosomes toward viral RNA messages. We reconstituted two in vitro reactions targeting a pre-translocation and a post-translocation state of the IAPV-IRES in the ribosome, allowing us to identify six structures using image processing classification methods. From these, we reconstructed the trajectory of IAPV-IRES from the early small subunit recruitment to the final post-translocated state in the ribosome. An early commitment of IRES/ribosome complexes for global pre-translocation mimicry explains the high efficiency observed for this IRES. Efforts directed toward fighting CCD by targeting the IAPV-IRES using RNA-interference technology are underway, and the structural framework presented here may assist in further refining these approaches.


Subject(s)
Biomimetics , Dicistroviridae/physiology , Internal Ribosome Entry Sites/genetics , Protein Biosynthesis , RNA, Transfer/genetics , RNA, Viral/genetics , Ribosomes/metabolism , Cryoelectron Microscopy , Dicistroviridae/ultrastructure , Humans , Models, Molecular , Nucleic Acid Conformation , RNA, Transfer/ultrastructure , Ribosomes/ultrastructure
12.
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
13.
Subcell Biochem ; 96: 433-450, 2021.
Article in English | MEDLINE | ID: mdl-33252739

ABSTRACT

Expansion segments (ES) are insertions of a few to hundreds of nucleotides at discrete locations on eukaryotic ribosomal RNA (rRNA) chains. Some cluster around 'hot spots' involved in translation regulation and some may participate in biogenesis. Whether ES play the same roles in different organisms is currently unclear, especially since their size may vary dramatically from one species to another and very little is known about their functions. Most likely, ES variation is linked to adaptation to a particular environment. In this chapter, we compare the interaction networks of ES from four kinetoplastid parasites, which have evolved in diverse insect vectors and mammalian hosts: Trypanosoma cruzi, Trypanosoma brucei, Leishmania donovani and Leishmania major. Here, we comparatively analyze ribosome structures from these representative kinetoplastids and ascertain meaningful structural differences from mammalian ribosomes. We base our analysis on sequence alignments and three-dimensional structures of 80S ribosomes solved by cryo-electron microscopy (cryo-EM). Striking differences in size are observed between ribosomes of different parasites, indicating that not all ES are expanded equally. Larger ES are not always matched by large surrounding ES or proteins extensions in their vicinity, a particularity that may lead to clues about their biological function. ES display different species-specific patterns of conservation, which underscore the density of their interaction network at the surface of the ribosome. Making sense of the conservation patterns of ES is part of a global effort to lay the basis for functional studies aimed at discovering unique kinetoplastid-specific sites suitable for therapeutic applications against these human and often animal pathogens.


Subject(s)
Kinetoplastida/genetics , RNA, Ribosomal/genetics , Ribosomes/metabolism , Animals , Cryoelectron Microscopy , Eukaryotic Cells/metabolism , Humans , Kinetoplastida/pathogenicity , Ribosomes/chemistry , Ribosomes/ultrastructure
14.
Biophys J ; 120(20): 4307-4311, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34509504

ABSTRACT

This article bemoans the demise of truly modular open-source image processing systems, such as SPIDER, in recent years' development of tools for three-dimensional reconstruction in cryo-electron microscopy. Instead, today's users have to rely on the functionality of software systems that have little or no transparency. As a consequence, users of such packages no longer gain a conceptual understanding and intuitive grasp of the analytical routes leading from the stream of input data to the final density map. Possible remedies of this situation with free software are discussed.


Subject(s)
Image Processing, Computer-Assisted , Software , Cryoelectron Microscopy , Imaging, Three-Dimensional
15.
Proteomics ; 21(2): e2000125, 2021 01.
Article in English | MEDLINE | ID: mdl-33007145

ABSTRACT

The role of the ribosome in the regulation of gene expression has come into increased focus. It is proposed that ribosomes are catalytic engines capable of changing their protein composition in response to environmental stimuli. Time-resolved cryo-electron microscopy (cryo-EM) techniques are employed to identify quantitative changes in the protein composition and structure of the Saccharomyces cerevisiae 80S ribosomes after shifting the carbon source from glucose to glycerol. Using cryo-EM combined with the computational classification approach, it is found that a fraction of the yeast cells' 80S ribosomes lack ribosomal proteins at the entrance and exit sites for tRNAs, including uL16(RPL10), eS1(RPS1), uS11(RPS14A/B), and eS26(RPS26A/B). This fraction increased after a change from glucose to glycerol medium. The quantitative structural analysis supports the hypothesis that ribosomes are dynamic complexes that alter their composition in response to changes in growth or environmental conditions.


Subject(s)
Saccharomyces cerevisiae , Carbon , Cryoelectron Microscopy , Ribosomal Proteins , Ribosomes , Saccharomyces cerevisiae Proteins
16.
Biochemistry ; 60(46): 3449-3451, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34077195

ABSTRACT

Single-particle cryogenic electron microscopy (cryo-EM), whose full power was not realized until the advent of powerful detectors in 2012, has a unique position as a method of structure determination as it is capable of providing information about not only the structure but also the dynamical features of biomolecules. This information is of special importance in understanding virus-host interaction and explains the crucial role of cryo-EM in the efforts to find vaccinations and cures for pandemics the world has experienced in the past decade.


Subject(s)
Cryoelectron Microscopy , Host Microbial Interactions , Single Molecule Imaging , Coronavirus Infections/epidemiology , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Dengue/epidemiology , Dengue/prevention & control , Dengue/virology , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Humans , Pandemics/prevention & control , Viral Vaccines/administration & dosage , Zika Virus Infection/epidemiology , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
17.
EMBO J ; 36(7): 854-868, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28179369

ABSTRACT

During ribosome biogenesis in eukaryotes, nascent subunits are exported to the cytoplasm in a functionally inactive state. 60S subunits are activated through a series of cytoplasmic maturation events. The last known events in the cytoplasm are the release of Tif6 by Efl1 and Sdo1 and the release of the export adapter, Nmd3, by the GTPase Lsg1. Here, we have used cryo-electron microscopy to determine the structure of the 60S subunit bound by Nmd3, Lsg1, and Tif6. We find that a central domain of Nmd3 mimics the translation elongation factor eIF5A, inserting into the E site of the ribosome and pulling the L1 stalk into a closed position. Additional domains occupy the P site and extend toward the sarcin-ricin loop to interact with Tif6. Nmd3 and Lsg1 together embrace helix 69 of the B2a intersubunit bridge, inducing base flipping that we suggest may activate the GTPase activity of Lsg1.


Subject(s)
GTP-Binding Proteins/metabolism , Organelle Biogenesis , RNA-Binding Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cryoelectron Microscopy , GTP-Binding Proteins/chemistry , RNA-Binding Proteins/chemistry , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/ultrastructure , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry
18.
Nature ; 525(7570): 491-5, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26344199

ABSTRACT

During eukaryotic translation initiation, 43S complexes, comprising a 40S ribosomal subunit, initiator transfer RNA and initiation factors (eIF) 2, 3, 1 and 1A, attach to the 5'-terminal region of messenger RNA and scan along it to the initiation codon. Scanning on structured mRNAs also requires the DExH-box protein DHX29. Mammalian eIF3 contains 13 subunits and participates in nearly all steps of translation initiation. Eight subunits having PCI (proteasome, COP9 signalosome, eIF3) or MPN (Mpr1, Pad1, amino-terminal) domains constitute the structural core of eIF3, to which five peripheral subunits are flexibly linked. Here we present a cryo-electron microscopy structure of eIF3 in the context of the DHX29-bound 43S complex, showing the PCI/MPN core at ∼6 Šresolution. It reveals the organization of the individual subunits and their interactions with components of the 43S complex. We were able to build near-complete polyalanine-level models of the eIF3 PCI/MPN core and of two peripheral subunits. The implications for understanding mRNA ribosomal attachment and scanning are discussed.


Subject(s)
Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-3/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Peptide Chain Initiation, Translational , Ribosomes/chemistry , Ribosomes/metabolism , Binding Sites , Codon, Initiator/genetics , Cryoelectron Microscopy , Eukaryotic Initiation Factor-2/chemistry , Eukaryotic Initiation Factor-2/metabolism , Humans , Models, Molecular , Peptide Initiation Factors/metabolism , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA Helicases/chemistry , RNA Helicases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer, Met/metabolism , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL