Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Wiley Interdiscip Rev RNA ; 14(5): e1792, 2023.
Article in English | MEDLINE | ID: mdl-37132456

ABSTRACT

Translation accuracy is one of the most critical factors for protein synthesis. It is regulated by the ribosome and its dynamic behavior, along with translation factors that direct ribosome rearrangements to make translation a uniform process. Earlier structural studies of the ribosome complex with arrested translation factors laid the foundation for an understanding of ribosome dynamics and the translation process as such. Recent technological advances in time-resolved and ensemble cryo-EM have made it possible to study translation in real time at high resolution. These methods provided a detailed view of translation in bacteria for all three phases: initiation, elongation, and termination. In this review, we focus on translation factors (in some cases GTP activation) and their ability to monitor and respond to ribosome organization to enable efficient and accurate translation. This article is categorized under: Translation > Ribosome Structure/Function Translation > Mechanisms.


Subject(s)
Ribosomes , Cryoelectron Microscopy/methods , Ribosomes/metabolism
2.
Mol Cell ; 82(21): 4049-4063.e6, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36182693

ABSTRACT

In animals and plants, Dicer enzymes collaborate with double-stranded RNA-binding domain (dsRBD) proteins to convert precursor-microRNAs (pre-miRNAs) into miRNA duplexes. We report six cryo-EM structures of Drosophila Dicer-1 that show how Dicer-1 and its partner Loqs­PB cooperate (1) before binding pre-miRNA, (2) after binding and in a catalytically competent state, (3) after nicking one arm of the pre-miRNA, and (4) following complete dicing and initial product release. Our reconstructions suggest that pre-miRNA binds a rare, open conformation of the Dicer­1⋅Loqs­PB heterodimer. The Dicer-1 dsRBD and three Loqs­PB dsRBDs form a tight belt around the pre-miRNA, distorting the RNA helix to place the scissile phosphodiester bonds in the RNase III active sites. Pre-miRNA cleavage shifts the dsRBDs and partially closes Dicer-1, which may promote product release. Our data suggest a model for how the Dicer­1⋅Loqs­PB complex affects a complete cycle of pre-miRNA recognition, stepwise endonuclease cleavage, and product release.


Subject(s)
Drosophila Proteins , MicroRNAs , Animals , Ribonuclease III/genetics , Ribonuclease III/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , RNA-Binding Proteins/metabolism , Drosophila/genetics , MicroRNAs/genetics , MicroRNAs/metabolism
3.
Nat Commun ; 13(1): 2776, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589706

ABSTRACT

Toxic dipeptide-repeat (DPR) proteins are produced from expanded G4C2 repeats in the C9ORF72 gene, the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Two DPR proteins, poly-PR and poly-GR, repress cellular translation but the molecular mechanism remains unknown. Here we show that poly-PR and poly-GR of ≥20 repeats inhibit the ribosome's peptidyl-transferase activity at nanomolar concentrations, comparable to specific translation inhibitors. High-resolution cryogenic electron microscopy (cryo-EM) reveals that poly-PR and poly-GR block the polypeptide tunnel of the ribosome, extending into the peptidyl-transferase center (PTC). Consistent with these findings, the macrolide erythromycin, which binds in the tunnel, competes with poly-PR and restores peptidyl-transferase activity. Our results demonstrate that strong and specific binding of poly-PR and poly-GR in the ribosomal tunnel blocks translation, revealing the structural basis of their toxicity in C9ORF72-ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Cryoelectron Microscopy , Dipeptides/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Humans , Proteins/genetics , Proteins/metabolism , Ribosomes/metabolism , Transferases
4.
Elife ; 112022 02 18.
Article in English | MEDLINE | ID: mdl-35179493

ABSTRACT

Sliding clamps are ring-shaped protein complexes that are integral to the DNA replication machinery of all life. Sliding clamps are opened and installed onto DNA by clamp loader AAA+ ATPase complexes. However, how a clamp loader opens and closes the sliding clamp around DNA is still unknown. Here, we describe structures of the Saccharomyces cerevisiae clamp loader Replication Factor C (RFC) bound to its cognate sliding clamp Proliferating Cell Nuclear Antigen (PCNA) en route to successful loading. RFC first binds to PCNA in a dynamic, closed conformation that blocks both ATPase activity and DNA binding. RFC then opens the PCNA ring through a large-scale 'crab-claw' expansion of both RFC and PCNA that explains how RFC prefers initial binding of PCNA over DNA. Next, the open RFC:PCNA complex binds DNA and interrogates the primer-template junction using a surprising base-flipping mechanism. Our structures indicate that initial PCNA opening and subsequent closure around DNA do not require ATP hydrolysis, but are driven by binding energy. ATP hydrolysis, which is necessary for RFC release, is triggered by interactions with both PCNA and DNA, explaining RFC's switch-like ATPase activity. Our work reveals how a AAA+ machine undergoes dramatic conformational changes for achieving binding preference and substrate remodeling.


Subject(s)
DNA Replication , Saccharomyces cerevisiae , ATPases Associated with Diverse Cellular Activities/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , DNA/metabolism , DNA-Directed DNA Polymerase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Replication Protein C/chemistry , Replication Protein C/genetics , Replication Protein C/metabolism , Saccharomyces cerevisiae/genetics
5.
Nat Commun ; 12(1): 7236, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903725

ABSTRACT

During translation, a conserved GTPase elongation factor-EF-G in bacteria or eEF2 in eukaryotes-translocates tRNA and mRNA through the ribosome. EF-G has been proposed to act as a flexible motor that propels tRNA and mRNA movement, as a rigid pawl that biases unidirectional translocation resulting from ribosome rearrangements, or by various combinations of motor- and pawl-like mechanisms. Using time-resolved cryo-EM, we visualized GTP-catalyzed translocation without inhibitors, capturing elusive structures of ribosome•EF-G intermediates at near-atomic resolution. Prior to translocation, EF-G binds near peptidyl-tRNA, while the rotated 30S subunit stabilizes the EF-G GTPase center. Reverse 30S rotation releases Pi and translocates peptidyl-tRNA and EF-G by ~20 Å. An additional 4-Å translocation initiates EF-G dissociation from a transient ribosome state with highly swiveled 30S head. The structures visualize how nearly rigid EF-G rectifies inherent and spontaneous ribosomal dynamics into tRNA-mRNA translocation, whereas GTP hydrolysis and Pi release drive EF-G dissociation.


Subject(s)
Cryoelectron Microscopy , Guanosine Triphosphate/chemistry , Peptide Elongation Factor G/chemistry , Ribosomes/chemistry , Escherichia coli/chemistry , Escherichia coli/metabolism , Guanosine Triphosphate/metabolism , Peptide Elongation Factor G/metabolism , Phosphates/metabolism , Protein Binding , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Transfer/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosome Subunits, Small, Bacterial/chemistry , Ribosome Subunits, Small, Bacterial/metabolism , Ribosomes/metabolism
6.
Nat Commun ; 12(1): 4644, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34330903

ABSTRACT

Frameshifting of mRNA during translation provides a strategy to expand the coding repertoire of cells and viruses. How and where in the elongation cycle +1-frameshifting occurs remains poorly understood. We describe seven ~3.5-Å-resolution cryo-EM structures of 70S ribosome complexes, allowing visualization of elongation and translocation by the GTPase elongation factor G (EF-G). Four structures with a + 1-frameshifting-prone mRNA reveal that frameshifting takes place during translocation of tRNA and mRNA. Prior to EF-G binding, the pre-translocation complex features an in-frame tRNA-mRNA pairing in the A site. In the partially translocated structure with EF-G•GDPCP, the tRNA shifts to the +1-frame near the P site, rendering the freed mRNA base to bulge between the P and E sites and to stack on the 16S rRNA nucleotide G926. The ribosome remains frameshifted in the nearly post-translocation state. Our findings demonstrate that the ribosome and EF-G cooperate to induce +1 frameshifting during tRNA-mRNA translocation.


Subject(s)
Frameshifting, Ribosomal/genetics , Peptide Chain Elongation, Translational/genetics , Peptide Elongation Factor G/genetics , RNA, Messenger/genetics , RNA, Transfer/genetics , Ribosomes/genetics , Biocatalysis , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Nucleic Acid Conformation , Peptide Elongation Factor G/chemistry , Peptide Elongation Factor G/metabolism , Protein Conformation , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomes/metabolism , Ribosomes/ultrastructure , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
7.
Nat Commun ; 11(1): 5552, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33144582

ABSTRACT

Ribosomes stalled during translation must be rescued to replenish the pool of translation-competent ribosomal subunits. Bacterial alternative rescue factor B (ArfB) releases nascent peptides from ribosomes stalled on mRNAs truncated at the A site, allowing ribosome recycling. Prior structural work revealed that ArfB recognizes such ribosomes by inserting its C-terminal α-helix into the vacant mRNA tunnel. In this work, we report that ArfB can efficiently recognize a wider range of mRNA substrates, including longer mRNAs that extend beyond the A-site codon. Single-particle cryo-EM unveils that ArfB employs two modes of function depending on the mRNA length. ArfB acts as a monomer to accommodate a shorter mRNA in the ribosomal A site. By contrast, longer mRNAs are displaced from the mRNA tunnel by more than 20 Å and are stabilized in the intersubunit space by dimeric ArfB. Uncovering distinct modes of ArfB function resolves conflicting biochemical and structural studies, and may lead to re-examination of other ribosome rescue pathways, whose functions depend on mRNA lengths.


Subject(s)
Escherichia coli Proteins/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , Biocatalysis , Dimerization , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/ultrastructure , Models, Biological , Protein Conformation , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/ultrastructure , Ribosome Subunits/metabolism , Ribosomes/ultrastructure
8.
Nature ; 584(7822): 640-645, 2020 08.
Article in English | MEDLINE | ID: mdl-32612237

ABSTRACT

Ribosomes accurately decode mRNA by proofreading each aminoacyl-tRNA that is delivered by the elongation factor EF-Tu1. To understand the molecular mechanism of this proofreading step it is necessary to visualize GTP-catalysed elongation, which has remained a challenge2-4. Here we use time-resolved cryogenic electron microscopy to reveal 33 ribosomal states after the delivery of aminoacyl-tRNA by EF-Tu•GTP. Instead of locking cognate tRNA upon initial recognition, the ribosomal decoding centre dynamically monitors codon-anticodon interactions before and after GTP hydrolysis. GTP hydrolysis enables the GTPase domain of EF-Tu to extend away, releasing EF-Tu from tRNA. The 30S subunit then locks cognate tRNA in the decoding centre and rotates, enabling the tRNA to bypass 50S protrusions during accommodation into the peptidyl transferase centre. By contrast, the decoding centre fails to lock near-cognate tRNA, enabling the dissociation of near-cognate tRNA both during initial selection (before GTP hydrolysis) and proofreading (after GTP hydrolysis). These findings reveal structural similarity between ribosomes in initial selection states5,6 and in proofreading states, which together govern the efficient rejection of incorrect tRNA.


Subject(s)
Cryoelectron Microscopy , Guanosine Triphosphate/metabolism , Peptide Elongation Factor Tu/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ribosomes/metabolism , Ribosomes/ultrastructure , Escherichia coli , GTP Phosphohydrolases/metabolism , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Hydrolysis , Models, Molecular , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/chemistry , RNA, Transfer/ultrastructure , Ribosomes/chemistry , Rotation
9.
Int J Biol Macromol ; 152: 1113-1124, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-31751748

ABSTRACT

Burkholderia pseudomallei and Chromobacterium violaceum are bacteria of tropical and subtropical soil and water that occasionally cause fatal infections in humans and animals. Microbial lectins mediate the adhesion of organisms to host cells, which is the first phase in the development of infection. Here we report the discovery of two novel lectins from the above-mentioned bacteria - BP39L and CV39L. The crystal structures revealed that the lectins possess a seven-bladed ß-propeller fold. Functional studies conducted on a series of oligo- and polysaccharides confirmed the preference of BP39L for mannosylated saccharides and CV39L for rather more complex polysaccharides with a monosaccharide preference for ß-l-fucose. The presented data indicate that the proteins belong to a currently unknown family of lectins.


Subject(s)
Bacterial Proteins/metabolism , Burkholderia pseudomallei/metabolism , Chromobacterium/metabolism , Lectins/metabolism , Animals , Fucose/metabolism , Humans , Monosaccharides/metabolism , Polysaccharides/metabolism
10.
Nat Commun ; 10(1): 4471, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578335

ABSTRACT

The capsids of double-stranded DNA viruses protect the viral genome from the harsh extracellular environment, while maintaining stability against the high internal pressure of packaged DNA. To elucidate how capsids maintain stability in an extreme environment, we use cryoelectron microscopy to determine the capsid structure of thermostable phage P74-26 to 2.8-Å resolution. We find P74-26 capsids exhibit an overall architecture very similar to those of other tailed bacteriophages, allowing us to directly compare structures to derive the structural basis for enhanced stability. Our structure reveals lasso-like interactions that appear to function like catch bonds. This architecture allows the capsid to expand during genome packaging, yet maintain structural stability. The P74-26 capsid has T = 7 geometry despite being twice as large as mesophilic homologs. Capsid capacity is increased with a larger, flatter major capsid protein. Given these results, we predict decreased icosahedral complexity (i.e. T ≤ 7) leads to a more stable capsid assembly.


Subject(s)
Bacteriophages/genetics , Capsid Proteins/genetics , Capsid/metabolism , Genome, Viral/genetics , Genomic Instability/genetics , Virion/genetics , Bacteriophages/metabolism , Bacteriophages/ultrastructure , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , DNA, Viral/chemistry , DNA, Viral/genetics , DNA, Viral/ultrastructure , Hot Temperature , Models, Molecular , Thermus thermophilus/virology , Virion/chemistry , Virion/ultrastructure , Virus Assembly/genetics
11.
Elife ; 82019 09 12.
Article in English | MEDLINE | ID: mdl-31513010

ABSTRACT

Protein synthesis ends when a ribosome reaches an mRNA stop codon. Release factors (RFs) decode the stop codon, hydrolyze peptidyl-tRNA to release the nascent protein, and then dissociate to allow ribosome recycling. To visualize termination by RF2, we resolved a cryo-EM ensemble of E. coli 70S•RF2 structures at up to 3.3 Å in a single sample. Five structures suggest a highly dynamic termination pathway. Upon peptidyl-tRNA hydrolysis, the CCA end of deacyl-tRNA departs from the peptidyl transferase center. The catalytic GGQ loop of RF2 is rearranged into a long ß-hairpin that plugs the peptide tunnel, biasing a nascent protein toward the ribosome exit. Ribosomal intersubunit rotation destabilizes the catalytic RF2 domain on the 50S subunit and disassembles the central intersubunit bridge B2a, resulting in RF2 departure. Our structures visualize how local rearrangements and spontaneous inter-subunit rotation poise the newly-made protein and RF2 to dissociate in preparation for ribosome recycling.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Peptide Chain Termination, Translational , Peptide Termination Factors/metabolism , Ribosomes/metabolism , Cryoelectron Microscopy , Escherichia coli Proteins/chemistry , Peptide Termination Factors/chemistry , Ribosomes/chemistry
12.
J Biol Chem ; 293(32): 12472-12479, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29941456

ABSTRACT

Accurate translation termination by release factors (RFs) is critical for the integrity of cellular proteomes. Premature termination on sense codons, for example, results in truncated proteins, whose accumulation could be detrimental to the cell. Nevertheless, some sense codons are prone to triggering premature termination, but the structural basis for this is unclear. To investigate premature termination, we determined a cryo-EM structure of the Escherichia coli 70S ribosome bound with RF1 in response to a UAU (Tyr) sense codon. The structure reveals that RF1 recognizes a UAU codon similarly to a UAG stop codon, suggesting that sense codons induce premature termination because they structurally mimic a stop codon. Hydrophobic interaction between the nucleobase of U3 (the third position of the UAU codon) and conserved Ile-196 in RF1 is important for misreading the UAU codon. Analyses of RNA binding in ribonucleoprotein complexes or by amino acids reveal that Ile-U packing is a frequent protein-RNA-binding motif with key functional implications. We discuss parallels with eukaryotic translation termination by the release factor eRF1.


Subject(s)
Codon, Terminator/metabolism , Codon/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Peptide Chain Termination, Translational , Peptide Termination Factors/metabolism , Ribosomes/metabolism , Codon/chemistry , Codon/genetics , Codon, Terminator/chemistry , Codon, Terminator/genetics , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Protein Conformation , Ribosomes/chemistry
13.
Proteins ; 86(9): 897-911, 2018 09.
Article in English | MEDLINE | ID: mdl-29722060

ABSTRACT

We report the characterization of the dimeric protein AB21 from Agaricus bisporus, one of the most commonly and widely consumed mushrooms in the world. The protein shares no significant sequence similarity with any protein of known function, and it is the first characterized member of its protein family. The coding sequence of the ab21 gene was determined and the protein was expressed in E. coli in a recombinant form. We demonstrated a high thermal and pH stability of AB21 and proved the weak affinity of the protein to divalent ions of some transition metals (nickel, zinc, cadmium, and cobalt). The reported crystallographic structure exhibits an interesting rod-like helical bundle fold with structural similarity to bacterial toxins of the ClyA superfamily. By immunostaining, we demonstrated an abundance of AB21 in the fruiting bodies of A. bisporus.


Subject(s)
Agaricus/chemistry , Bacterial Toxins/chemistry , Fungal Proteins/biosynthesis , Pore Forming Cytotoxic Proteins/biosynthesis , Cations, Divalent/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Protein Conformation , Protein Folding , Protein Stability , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Transition Elements/chemistry
14.
Elife ; 62017 10 13.
Article in English | MEDLINE | ID: mdl-29027901

ABSTRACT

In bacteria, mRNA transcription and translation are coupled to coordinate optimal gene expression and maintain genome stability. Coupling is thought to involve direct interactions between RNA polymerase (RNAP) and the translational machinery. We present cryo-EM structures of E. coli RNAP core bound to the small ribosomal 30S subunit. The complex is stable under cell-like ionic conditions, consistent with functional interaction between RNAP and the 30S subunit. The RNA exit tunnel of RNAP aligns with the Shine-Dalgarno-binding site of the 30S subunit. Ribosomal protein S1 forms a wall of the tunnel between RNAP and the 30S subunit, consistent with its role in directing mRNAs onto the ribosome. The nucleic-acid-binding cleft of RNAP samples distinct conformations, suggesting different functional states during transcription-translation coupling. The architecture of the 30S•RNAP complex provides a structural basis for co-localization of the transcriptional and translational machineries, and inform future mechanistic studies of coupled transcription and translation.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/enzymology , Ribosome Subunits, Small, Bacterial/chemistry , Ribosome Subunits, Small, Bacterial/metabolism , Cryoelectron Microscopy , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism
15.
J Biol Chem ; 292(42): 17525-17540, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28860196

ABSTRACT

Multistep phosphorelay (MSP) cascades mediate responses to a wide spectrum of stimuli, including plant hormonal signaling, but several aspects of MSP await elucidation. Here, we provide first insight into the key step of MSP-mediated phosphotransfer in a eukaryotic system, the phosphorylation of the receiver domain of the histidine kinase CYTOKININ-INDEPENDENT 1 (CKI1RD) from Arabidopsis thaliana We observed that the crystal structures of free, Mg2+-bound, and beryllofluoridated CKI1RD (a stable analogue of the labile phosphorylated form) were identical and similar to the active state of receiver domains of bacterial response regulators. However, the three CKI1RD variants exhibited different conformational dynamics in solution. NMR studies revealed that Mg2+ binding and beryllofluoridation alter the conformational equilibrium of the ß3-α3 loop close to the phosphorylation site. Mutations that perturbed the conformational behavior of the ß3-α3 loop while keeping the active-site aspartate intact resulted in suppression of CKI1 function. Mechanistically, homology modeling indicated that the ß3-α3 loop directly interacts with the ATP-binding site of the CKI1 histidine kinase domain. The functional relevance of the conformational dynamics observed in the ß3-α3 loop of CKI1RD was supported by a comparison with another A. thaliana histidine kinase, ETR1. In contrast to the highly dynamic ß3-α3 loop of CKI1RD, the corresponding loop of the ETR1 receiver domain (ETR1RD) exhibited little conformational exchange and adopted a different orientation in crystals. Biochemical data indicated that ETR1RD is involved in phosphorylation-independent signaling, implying a direct link between conformational behavior and the ability of eukaryotic receiver domains to participate in MSP.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/enzymology , Protein Kinases/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Crystallography, X-Ray , Nuclear Magnetic Resonance, Biomolecular , Protein Domains , Protein Kinases/genetics , Protein Structure, Secondary , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics
16.
PLoS Pathog ; 13(8): e1006564, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28806750

ABSTRACT

Photorhabdus asymbiotica is one of the three recognized species of the Photorhabdus genus, which consists of gram-negative bioluminescent bacteria belonging to the family Morganellaceae. These bacteria live in a symbiotic relationship with nematodes from the genus Heterorhabditis, together forming a complex that is highly pathogenic for insects. Unlike other Photorhabdus species, which are strictly entomopathogenic, P. asymbiotica is unique in its ability to act as an emerging human pathogen. Analysis of the P. asymbiotica genome identified a novel fucose-binding lectin designated PHL with a strong sequence similarity to the recently described P. luminescens lectin PLL. Recombinant PHL exhibited high affinity for fucosylated carbohydrates and the unusual disaccharide 3,6-O-Me2-Glcß1-4(2,3-O-Me2)Rhaα-O-(p-C6H4)-OCH2CH2NH2 from Mycobacterium leprae. Based on its crystal structure, PHL forms a seven-bladed ß-propeller assembling into a homo-dimer with an inter-subunit disulfide bridge. Investigating complexes with different ligands revealed the existence of two sets of binding sites per monomer-the first type prefers l-fucose and its derivatives, whereas the second type can bind d-galactose. Based on the sequence analysis, PHL could contain up to twelve binding sites per monomer. PHL was shown to interact with all types of red blood cells and insect haemocytes. Interestingly, PHL inhibited the production of reactive oxygen species induced by zymosan A in human blood and antimicrobial activity both in human blood, serum and insect haemolymph. Concurrently, PHL increased the constitutive level of oxidants in the blood and induced melanisation in haemolymph. Our results suggest that PHL might play a crucial role in the interaction of P. asymbiotica with both human and insect hosts.


Subject(s)
Bacterial Proteins/immunology , Host-Pathogen Interactions/immunology , Lectins/immunology , Photorhabdus/immunology , Animals , Bacterial Proteins/genetics , Base Sequence , Crystallography, X-Ray , Humans , Lectins/chemistry , Lectins/genetics , Molecular Sequence Data , Photorhabdus/genetics , Protein Conformation , Surface Plasmon Resonance
17.
Nature ; 546(7656): 113-117, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28538735

ABSTRACT

Gene translation depends on accurate decoding of mRNA, the structural mechanism of which remains poorly understood. Ribosomes decode mRNA codons by selecting cognate aminoacyl-tRNAs delivered by elongation factor Tu (EF-Tu). Here we present high-resolution structural ensembles of ribosomes with cognate or near-cognate aminoacyl-tRNAs delivered by EF-Tu. Both cognate and near-cognate tRNA anticodons explore the aminoacyl-tRNA-binding site (A site) of an open 30S subunit, while inactive EF-Tu is separated from the 50S subunit. A transient conformation of decoding-centre nucleotide G530 stabilizes the cognate codon-anticodon helix, initiating step-wise 'latching' of the decoding centre. The resulting closure of the 30S subunit docks EF-Tu at the sarcin-ricin loop of the 50S subunit, activating EF-Tu for GTP hydrolysis and enabling accommodation of the aminoacyl-tRNA. By contrast, near-cognate complexes fail to induce the G530 latch, thus favouring open 30S pre-accommodation intermediates with inactive EF-Tu. This work reveals long-sought structural differences between the pre-accommodation of cognate and near-cognate tRNAs that elucidate the mechanism of accurate decoding.


Subject(s)
Cryoelectron Microscopy , Protein Biosynthesis , Ribosomes/metabolism , Ribosomes/ultrastructure , Anticodon/chemistry , Anticodon/genetics , Anticodon/ultrastructure , Codon/chemistry , Codon/genetics , Codon/ultrastructure , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/ultrastructure , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/ultrastructure , Guanosine Triphosphate/metabolism , Hydrolysis , Models, Molecular , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factor Tu/ultrastructure , Protein Domains , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , RNA, Ribosomal, 16S/ultrastructure , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Amino Acyl/ultrastructure , Ribosome Subunits/chemistry , Ribosome Subunits/metabolism , Ribosome Subunits/ultrastructure , Ribosomes/chemistry
18.
Elife ; 62017 03 16.
Article in English | MEDLINE | ID: mdl-28300532

ABSTRACT

ArfA rescues ribosomes stalled on truncated mRNAs by recruiting release factor RF2, which normally binds stop codons to catalyze peptide release. We report two 3.2 Šresolution cryo-EM structures - determined from a single sample - of the 70S ribosome with ArfA•RF2 in the A site. In both states, the ArfA C-terminus occupies the mRNA tunnel downstream of the A site. One state contains a compact inactive RF2 conformation. Ordering of the ArfA N-terminus in the second state rearranges RF2 into an extended conformation that docks the catalytic GGQ motif into the peptidyl-transferase center. Our work thus reveals the structural dynamics of ribosome rescue. The structures demonstrate how ArfA 'senses' the vacant mRNA tunnel and activates RF2 to mediate peptide release without a stop codon, allowing stalled ribosomes to be recycled.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , Peptide Termination Factors/metabolism , Peptide Termination Factors/ultrastructure , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/ultrastructure , Ribosomes/metabolism , Ribosomes/ultrastructure , Cryoelectron Microscopy , Protein Binding
19.
J Biol Chem ; 291(48): 25032-25049, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27758853

ABSTRACT

Photorhabdus luminescens is known for its symbiosis with the entomopathogenic nematode Heterorhabditis bacteriophora and its pathogenicity toward insect larvae. A hypothetical protein from P. luminescens was identified, purified from the native source, and characterized as an l-fucose-binding lectin, named P. luminescens lectin (PLL). Glycan array and biochemical characterization data revealed PLL to be specific toward l-fucose and the disaccharide glycan 3,6-O-Me2-Glcß1-4(2,3-O-Me2)Rhaα-O-(p-C6H4)-OCH2CH2NH2 PLL was discovered to be a homotetramer with an intersubunit disulfide bridge. The crystal structures of native and recombinant PLL revealed a seven-bladed ß-propeller fold creating seven putative fucose-binding sites per monomer. The crystal structure of the recombinant PLL·l-fucose complex confirmed that at least three sites were fucose-binding. Moreover, the crystal structures indicated that some of the other sites are masked either by the tetrameric nature of the lectin or by incorporation of the C terminus of the lectin into one of these sites. PLL exhibited an ability to bind to insect hemocytes and the cuticular surface of a nematode, H. bacteriophora.


Subject(s)
Bacterial Proteins/chemistry , Fucose/chemistry , Lectins/chemistry , Photorhabdus/chemistry , Bacterial Proteins/isolation & purification , Crystallography, X-Ray , Lectins/isolation & purification , Protein Domains , Protein Structure, Quaternary
20.
Proteins ; 84(5): 686-99, 2016 May.
Article in English | MEDLINE | ID: mdl-26879585

ABSTRACT

Sequence dependence of (13) C and (15) N chemical shifts in the receiver domain of CKI1 protein from Arabidopsis thaliana, CKI1RD , and its complexed form, CKI1RD •Mg(2+), was studied by means of MD/DFT calculations. MD simulations of a 20-ns production run length were performed. Nine explicitly hydrated structures of increasing complexity were explored, up to a 40-amino-acid structure. The size of the model necessary depended on the type of nucleus, the type of amino acid and its sequence neighbors, other spatially close amino acids, and the orientation of amino acid NH groups and their surface/interior position. Using models covering a 10 and a 15 Å environment of Mg(2+), a semi-quantitative agreement has been obtained between experiment and theory for the V67-I73 sequence. The influence of Mg(2+) binding was described better by the 15 Å as compared to the 10 Å model. Thirteen chemical shifts were analyzed in terms of the effect of Mg(2+) insertion and geometry preparation. The effect of geometry was significant and opposite in sign to the effect of Mg(2+) binding. The strongest individual effects were found for (15) N of D70, S74, and V68, where the electrostatics dominated; for (13) Cß of D69 and (15) N of K76, where the influences were equal, and for (13) Cα of F72 and (13) Cß of K76, where the geometry adjustment dominated. A partial correlation between dominant geometry influence and torsion angle shifts upon the coordination has been observed.


Subject(s)
Arabidopsis Proteins/chemistry , Carbon Isotopes/chemistry , Magnesium/chemistry , Nitrogen Isotopes/chemistry , Protein Kinases/chemistry , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...