Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Microbiol ; 119(5): 630-639, 2023 05.
Article in English | MEDLINE | ID: mdl-37024243

ABSTRACT

There are multiple RNA degradation mechanisms in eukaryotes, key among these is mRNA decapping, which requires the Dcp1-Dcp2 complex. Decapping is involved in various processes including nonsense-mediated decay (NMD), a process by which aberrant transcripts with a premature termination codon are targeted for translational repression and rapid decay. NMD is ubiquitous throughout eukaryotes and the key factors involved are highly conserved, although many differences have evolved. We investigated the role of Aspergillus nidulans decapping factors in NMD and found that they are not required, unlike Saccharomyces cerevisiae. Intriguingly, we also observed that the disruption of one of the decapping factors, Dcp1, leads to an aberrant ribosome profile. Importantly this was not shared by mutations disrupting Dcp2, the catalytic component of the decapping complex. The aberrant profile is associated with the accumulation of a high proportion of 25S rRNA degradation intermediates. We identified the location of three rRNA cleavage sites and show that a mutation targeted to disrupt the catalytic domain of Dcp2 partially suppresses the aberrant profile of Δdcp1 strains. This suggests that in the absence of Dcp1, cleaved ribosomal components accumulate and Dcp2 may be directly involved in mediating these cleavage events. We discuss the implications of this.


Subject(s)
Aspergillus nidulans , Saccharomyces cerevisiae Proteins , Aspergillus nidulans/genetics , Aspergillus nidulans/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Nonsense Mediated mRNA Decay , Ribosomes/genetics , Ribosomes/metabolism , Endoribonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism
2.
Mol Microbiol ; 115(2): 238-254, 2021 02.
Article in English | MEDLINE | ID: mdl-33047379

ABSTRACT

The role of post-transcriptional RNA modification is of growing interest. One example is the addition of non-templated uridine residues to the 3' end of transcripts. In mammalian systems, uridylation is integral to cell cycle control of histone mRNA levels. This regulatory mechanism is dependent on the nonsense-mediated decay (NMD) component, Upf1, which promotes histone mRNA uridylation and degradation in response to the arrest of DNA synthesis. We have identified a similar system in Aspergillus nidulans, where Upf1 is required for the regulation of histone mRNA levels. However, other NMD components are also implicated, distinguishing it from the mammalian system. As in human cells, 3' uridylation of histone mRNA is induced upon replication arrest. Disruption of this 3' tagging has a significant but limited effect on histone transcript regulation, consistent with multiple mechanisms acting to regulate mRNA levels. Interestingly, 3' end degraded transcripts are also subject to re-adenylation. Both mRNA pyrimidine tagging and re-adenylation are dependent on the same terminal-nucleotidyltransferases, CutA, and CutB, and we show this is consistent with the in vitro activities of both enzymes. Based on these data we argue that mRNA 3' tagging has diverse and distinct roles associated with transcript degradation, functionality and regulation.


Subject(s)
Aspergillus nidulans/genetics , Histones/genetics , RNA, Messenger/genetics , 3' Untranslated Regions/genetics , DNA Replication/physiology , Glutathione/analogs & derivatives , Glutathione/genetics , Glutathione/metabolism , Histones/metabolism , Nonsense Mediated mRNA Decay , RNA Helicases/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/physiology , RNA Stability , RNA, Messenger/metabolism , Trans-Activators/metabolism , Uridine/chemistry
3.
Nucleic Acids Res ; 45(4): 2179-2187, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27986852

ABSTRACT

In bacteria, the start site and the reading frame of the messenger RNA are selected by the small ribosomal subunit (30S) when the start codon, typically an AUG, is decoded in the P-site by the initiator tRNA in a process guided and controlled by three initiation factors. This process can be efficiently inhibited by GE81112, a natural tetrapeptide antibiotic that is highly specific toward bacteria. Here GE81112 was used to stabilize the 30S pre-initiation complex and obtain its structure by cryo-electron microscopy. The results obtained reveal the occurrence of changes in both the ribosome conformation and initiator tRNA position that may play a critical role in controlling translational fidelity. Furthermore, the structure highlights similarities with the early steps of initiation in eukaryotes suggesting that shared structural features guide initiation in all kingdoms of life.


Subject(s)
Peptide Chain Initiation, Translational , RNA, Messenger/genetics , RNA, Transfer, Met/genetics , Ribosome Subunits, Small, Bacterial/metabolism , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryotic Cells/metabolism , Models, Molecular , Molecular Conformation , Prokaryotic Initiation Factors/chemistry , Prokaryotic Initiation Factors/metabolism , Protein Biosynthesis/genetics , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Transfer, Met/chemistry , RNA, Transfer, Met/metabolism , Ribosome Subunits, Large, Bacterial/chemistry , Ribosome Subunits, Large, Bacterial/metabolism , Ribosome Subunits, Small, Bacterial/chemistry
4.
Nucleic Acids Res ; 45(11): 6945-6959, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28482099

ABSTRACT

During 30S ribosomal subunit biogenesis, assembly factors are believed to prevent accumulation of misfolded intermediate states of low free energy that slowly convert into mature 30S subunits, namely, kinetically trapped particles. Among the assembly factors, the circularly permuted GTPase, RsgA, plays a crucial role in the maturation of the 30S decoding center. Here, directed hydroxyl radical probing and single particle cryo-EM are employed to elucidate RsgA΄s mechanism of action. Our results show that RsgA destabilizes the 30S structure, including late binding r-proteins, providing a structural basis for avoiding kinetically trapped assembly intermediates. Moreover, RsgA exploits its distinct GTPase pocket and specific interactions with the 30S to coordinate GTPase activation with the maturation state of the 30S subunit. This coordination validates the architecture of the decoding center and facilitates the timely release of RsgA to control the progression of 30S biogenesis.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , GTP Phosphohydrolases/chemistry , Catalytic Domain , Cryoelectron Microscopy , Enzyme Activation , Escherichia coli Proteins/physiology , GTP Phosphohydrolases/physiology , Guanosine Triphosphate/chemistry , Hydrogen Bonding , Hydrolysis , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Ribosome Subunits, Small, Bacterial
5.
Proc Natl Acad Sci U S A ; 113(16): E2286-95, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27071098

ABSTRACT

In prokaryotic systems, the initiation phase of protein synthesis is governed by the presence of initiation factors that guide the transition of the small ribosomal subunit (30S) from an unlocked preinitiation complex (30S preIC) to a locked initiation complex (30SIC) upon the formation of a correct codon-anticodon interaction in the peptidyl (P) site. Biochemical and structural characterization of GE81112, a translational inhibitor specific for the initiation phase, indicates that the main mechanism of action of this antibiotic is to prevent P-site decoding by stabilizing the anticodon stem loop of the initiator tRNA in a distorted conformation. This distortion stalls initiation in the unlocked 30S preIC state characterized by tighter IF3 binding and a reduced association rate for the 50S subunit. At the structural level we observe that in the presence of GE81112 the h44/h45/h24a interface, which is part of the IF3 binding site and forms ribosomal intersubunit bridges, preferentially adopts a disengaged conformation. Accordingly, the findings reveal that the dynamic equilibrium between the disengaged and engaged conformations of the h44/h45/h24a interface regulates the progression of protein synthesis, acting as a molecular switch that senses and couples the 30S P-site decoding step of translation initiation to the transition from an unlocked preIC to a locked 30SIC state.


Subject(s)
Anti-Bacterial Agents/chemistry , Escherichia coli/chemistry , Peptide Chain Initiation, Translational , RNA, Bacterial/chemistry , RNA, Ribosomal, 16S/chemistry , RNA, Transfer/chemistry , Ribosome Subunits, Small, Bacterial/chemistry , Nucleic Acid Conformation
6.
Nucleic Acids Res ; 43(20): 10015-25, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26464437

ABSTRACT

Hygromycin A (HygA) binds to the large ribosomal subunit and inhibits its peptidyl transferase (PT) activity. The presented structural and biochemical data indicate that HygA does not interfere with the initial binding of aminoacyl-tRNA to the A site, but prevents its subsequent adjustment such that it fails to act as a substrate in the PT reaction. Structurally we demonstrate that HygA binds within the peptidyl transferase center (PTC) and induces a unique conformation. Specifically in its ribosomal binding site HygA would overlap and clash with aminoacyl-A76 ribose moiety and, therefore, its primary mode of action involves sterically restricting access of the incoming aminoacyl-tRNA to the PTC.


Subject(s)
Cinnamates/chemistry , Cinnamates/pharmacology , Hygromycin B/analogs & derivatives , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Ribosome Subunits, Large, Bacterial/chemistry , Ribosome Subunits, Large, Bacterial/drug effects , Binding Sites , Cinnamates/metabolism , Crystallography, X-Ray , Hygromycin B/chemistry , Hygromycin B/metabolism , Hygromycin B/pharmacology , Models, Molecular , Peptidyl Transferases/chemistry , Peptidyl Transferases/drug effects , Protein Synthesis Inhibitors/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosome Subunits, Large, Bacterial/enzymology , Ribosome Subunits, Large, Bacterial/metabolism
7.
Nature ; 468(7324): 713-6, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21124459

ABSTRACT

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


Subject(s)
Movement , RNA, Transfer/metabolism , Ribosome Subunits, Small, Bacterial/chemistry , Ribosome Subunits, Small, Bacterial/metabolism , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Models, Molecular , Peptide Elongation Factor G/chemistry , Peptide Elongation Factor G/metabolism , Protein Biosynthesis , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Transfer/chemistry , RNA, Transfer/ultrastructure , Ribosome Subunits, Small, Bacterial/ultrastructure , Thermus thermophilus/chemistry
8.
Mol Cell ; 30(1): 26-38, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18406324

ABSTRACT

The thiopeptide class of antibiotics targets the GTPase-associated center (GAC) of the ribosome to inhibit translation factor function. Using X-ray crystallography, we have determined the binding sites of thiostrepton (Thio), nosiheptide (Nosi), and micrococcin (Micro), on the Deinococcus radiodurans large ribosomal subunit. The thiopeptides, by binding within a cleft located between the ribosomal protein L11 and helices 43 and 44 of the 23S rRNA, overlap with the position of domain V of EF-G, thus explaining how this class of drugs perturbs translation factor binding to the ribosome. The presence of Micro leads to additional density for the C-terminal domain (CTD) of L7, adjacent to and interacting with L11. The results suggest that L11 acts as a molecular switch to control L7 binding and plays a pivotal role in positioning one L7-CTD monomer on the G' subdomain of EF-G to regulate EF-G turnover during protein synthesis.


Subject(s)
Bacteriocins , Gene Expression Regulation , Peptides , Protein Biosynthesis , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomes , Thiostrepton , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacteriocins/chemistry , Bacteriocins/metabolism , Binding Sites , Crystallography, X-Ray , Deinococcus/chemistry , Deinococcus/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Peptides/chemistry , Peptides/metabolism , Protein Structure, Tertiary , Ribosomal Proteins/genetics , Ribosomes/chemistry , Ribosomes/metabolism , Thiazoles/chemistry , Thiazoles/metabolism , Thiostrepton/chemistry , Thiostrepton/metabolism
9.
Antimicrob Agents Chemother ; 59(5): 2849-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25753625

ABSTRACT

Although both tetracycline and tigecycline inhibit protein synthesis by sterically hindering the binding of tRNA to the ribosomal A site, tigecycline shows increased efficacy in both in vitro and in vivo activity assays and escapes the most common resistance mechanisms associated with the tetracycline class of antibiotics. These differences in activities are attributed to the tert-butyl-glycylamido side chain found in tigecycline. Our structural analysis by X-ray crystallography shows that tigecycline binds the bacterial 30S ribosomal subunit with its tail in an extended conformation and makes extensive interactions with the 16S rRNA nucleotide C1054. These interactions restrict the mobility of C1054 and contribute to the antimicrobial activity of tigecycline, including its resistance to the ribosomal protection proteins.


Subject(s)
Minocycline/analogs & derivatives , Ribosomes/metabolism , Crystallography, X-Ray , Minocycline/metabolism , Minocycline/pharmacology , Protein Binding , Protein Structure, Secondary , RNA, Ribosomal, 16S/metabolism , Thermus thermophilus/drug effects , Thermus thermophilus/metabolism , Tigecycline
10.
EMBO J ; 28(6): 755-65, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19229291

ABSTRACT

We have used single-particle reconstruction in cryo-electron microscopy to determine a structure of the Thermus thermophilus ribosome in which the ternary complex of elongation factor Tu (EF-Tu), tRNA and guanine nucleotide has been trapped on the ribosome using the antibiotic kirromycin. This represents the state in the decoding process just after codon recognition by tRNA and the resulting GTP hydrolysis by EF-Tu, but before the release of EF-Tu from the ribosome. Progress in sample purification and image processing made it possible to reach a resolution of 6.4 A. Secondary structure elements in tRNA, EF-Tu and the ribosome, and even GDP and kirromycin, could all be visualized directly. The structure reveals a complex conformational rearrangement of the tRNA in the A/T state and the interactions with the functionally important switch regions of EF-Tu crucial to GTP hydrolysis. Thus, the structure provides insights into the molecular mechanism of signalling codon recognition from the decoding centre of the 30S subunit to the GTPase centre of EF-Tu.


Subject(s)
Peptide Elongation Factor Tu/metabolism , Ribosomes/enzymology , Thermus thermophilus/enzymology , Cryoelectron Microscopy , Enzyme Activation , Guanosine Diphosphate/chemistry , Models, Molecular , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/ultrastructure , Protein Structure, Secondary , Pyridones/chemistry , RNA, Transfer/chemistry , RNA, Transfer/ultrastructure , Ribosomes/chemistry , Ribosomes/ultrastructure , Static Electricity
11.
J Biomol NMR ; 56(2): 85-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23689811

ABSTRACT

The impact of Nuclear Magnetic Resonance (NMR) on studies of large macromolecular complexes hinges on improvements in sensitivity and resolution. Dynamic nuclear polarization (DNP) in the solid state can offer improved sensitivity, provided sample preparation is optimized to preserve spectral resolution. For a few nanomoles of intact ribosomes and an 800 kDa ribosomal complex we demonstrate that the combination of DNP and magic-angle spinning NMR (MAS-NMR) allows one to overcome current sensitivity limitations so that homo- and heteronuclear (13)C and (15)N NMR correlation spectra can be recorded. Ribosome particles, directly pelleted and frozen into an NMR rotor, yield DNP signal enhancements on the order of ~25-fold and spectra that exhibit narrow linewidths, suitable for obtaining site-specific information. We anticipate that the same approach is applicable to other high molecular weight complexes.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Ribosomes/chemistry , Freezing , Models, Molecular , Molecular Conformation , Nuclear Magnetic Resonance, Biomolecular/methods
12.
Nat Struct Mol Biol ; 15(9): 910-5, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19172743

ABSTRACT

EF4 (LepA) is an almost universally conserved translational GTPase in eubacteria. It seems to be essential under environmental stress conditions and has previously been shown to back-translocate the tRNAs on the ribosome, thereby reverting the canonical translocation reaction. In the current work, EF4 was directly visualized in the process of back-translocating tRNAs by single-particle cryo-EM. Using flexible fitting methods, we built a model of ribosome-bound EF4 based on the cryo-EM map and a recently published unbound EF4 X-ray structure. The cryo-EM map establishes EF4 as a noncanonical elongation factor that interacts not only with the elongating ribosome, but also with the back-translocated tRNA in the A-site region, which is present in a previously unseen, intermediate state and deviates markedly from the position of a canonical A-tRNA. Our results, therefore, provide insight into the underlying structural principles governing back-translocation.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/metabolism , Biological Transport, Active , Escherichia coli/metabolism , Guanosine Triphosphate/metabolism , Hydrolysis , Macromolecular Substances , Models, Molecular , Peptide Initiation Factors
13.
Nat Microbiol ; 8(10): 1834-1845, 2023 10.
Article in English | MEDLINE | ID: mdl-37709902

ABSTRACT

Translational control is an essential process for the cell to adapt to varying physiological or environmental conditions. To survive adverse conditions such as low nutrient levels, translation can be shut down almost entirely by inhibiting ribosomal function. Here we investigated eukaryotic hibernating ribosomes from the microsporidian parasite Spraguea lophii in situ by a combination of electron cryo-tomography and single-particle electron cryo-microscopy. We show that microsporidian spores contain hibernating ribosomes that are locked in a dimeric (100S) state, which is formed by a unique dimerization mechanism involving the beak region. The ribosomes within the dimer are fully assembled, suggesting that they are ready to be activated once the host cell is invaded. This study provides structural evidence for dimerization acting as a mechanism for ribosomal hibernation in microsporidia, and therefore demonstrates that eukaryotes utilize this mechanism in translational control.


Subject(s)
Microsporidia , Animals , Cryoelectron Microscopy , Spores , Dimerization , Eukaryota , Ribosomes
14.
mBio ; 14(4): e0041423, 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37409813

ABSTRACT

Invasive aspergillosis is one of the most serious clinical invasive fungal infections, resulting in a high case fatality rate among immunocompromised patients. The disease is caused by saprophytic molds in the genus Aspergillus, including Aspergillus fumigatus, the most significant pathogenic species. The fungal cell wall, an essential structure mainly composed of glucan, chitin, galactomannan, and galactosaminogalactan, represents an important target for the development of antifungal drugs. UDP (uridine diphosphate)-glucose pyrophosphorylase (UGP) is a central enzyme in the metabolism of carbohydrates that catalyzes the biosynthesis of UDP-glucose, a key precursor of fungal cell wall polysaccharides. Here, we demonstrate that the function of UGP is vital for Aspergillus nidulans (AnUGP). To understand the molecular basis of AnUGP function, we describe a cryoEM structure (global resolution of 3.5 Å for the locally refined subunit and 4 Å for the octameric complex) of a native AnUGP. The structure reveals an octameric architecture with each subunit comprising an N-terminal α-helical domain, a central catalytic glycosyltransferase A-like (GT-A-like) domain, and a C-terminal (CT) left-handed ß-helix oligomerization domain. AnUGP displays unprecedented conformational variability between the CT oligomerization domain and the central GT-A-like catalytic domain. In combination with activity measurements and bioinformatics analysis, we unveil the molecular mechanism of substrate recognition and specificity for AnUGP. Altogether, our study not only contributes to understanding the molecular mechanism of catalysis/regulation of an important class of enzymes but also provides the genetic, biochemical, and structural groundwork for the future exploitation of UGP as a potential antifungal target. IMPORTANCE Fungi cause diverse diseases in humans, ranging from allergic syndromes to life-threatening invasive diseases, together affecting more than a billion people worldwide. Increasing drug resistance in Aspergillus species represents an emerging global health threat, making the design of antifungals with novel mechanisms of action a worldwide priority. The cryoEM structure of UDP (uridine diphosphate)-glucose pyrophosphorylase (UGP) from the filamentous fungus Aspergillus nidulans reveals an octameric architecture displaying unprecedented conformational variability between the C-terminal oligomerization domain and the central glycosyltransferase A-like catalytic domain in the individual protomers. While the active site and oligomerization interfaces are more highly conserved, these dynamic interfaces include motifs restricted to specific clades of filamentous fungi. Functional study of these motifs could lead to the definition of new targets for antifungals inhibiting UGP activity and, thus, the architecture of the cell wall of filamentous fungal pathogens.

15.
Front Microbiol ; 13: 870938, 2022.
Article in English | MEDLINE | ID: mdl-35495643

ABSTRACT

Two years after its emergence, the coronavirus disease-2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remains difficult to control despite the availability of several vaccines. The extensively glycosylated SARS-CoV-2 spike (S) protein, which mediates host cell entry by binding to the angiotensin converting enzyme 2 (ACE2) through its receptor binding domain (RBD), is the major target of neutralizing antibodies. Like to many other viral fusion proteins, the SARS-CoV-2 spike protein utilizes a glycan shield to thwart the host immune response. To grasp the influence of chemical signatures on carbohydrate mobility and reconcile the cryo-EM density of specific glycans we combined our cryo-EM map of the S ectodomain to 4.1 Å resolution, reconstructed from a limited number of particles, and all-atom molecular dynamics simulations. Chemical modifications modeled on representative glycans (defucosylation, sialylation and addition of terminal LacNAc units) show no significant influence on either protein shielding or glycan flexibility. By estimating at selected sites the local correlation between the full density map and atomic model-based maps derived from molecular dynamics simulations, we provide insight into the geometries of the α-Man-(1→3)-[α-Man-(1→6)-]-ß-Man-(1→4)-ß-GlcNAc(1→4)-ß-GlcNAc core common to all N-glycosylation sites.

16.
Nat Struct Mol Biol ; 13(12): 1092-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17115051

ABSTRACT

Internal ribosome entry sites (IRESs) facilitate an alternative, end-independent pathway of translation initiation. A particular family of dicistroviral IRESs can assemble elongation-competent 80S ribosomal complexes in the absence of canonical initiation factors and initiator transfer RNA. We present here a cryo-EM reconstruction of a dicistroviral IRES bound to the 80S ribosome. The resolution of the cryo-EM reconstruction, in the subnanometer range, allowed the molecular structure of the complete IRES in its active, ribosome-bound state to be solved. The structure, harboring three pseudoknot-containing domains, each with a specific functional role, shows how defined elements of the IRES emerge from a compactly folded core and interact with the key ribosomal components that form the A, P and E sites, where tRNAs normally bind. Our results exemplify the molecular strategy for recruitment of an IRES and reveal the dynamic features necessary for internal initiation.


Subject(s)
Gryllidae/virology , RNA Viruses/genetics , RNA, Viral/chemistry , RNA, Viral/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Animals , Base Sequence , Cryoelectron Microscopy , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Paralysis , Protein Binding , Protein Structure, Tertiary , RNA, Viral/ultrastructure , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomes/ultrastructure , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Structural Homology, Protein
17.
Proc Natl Acad Sci U S A ; 105(36): 13339-44, 2008 Sep 09.
Article in English | MEDLINE | ID: mdl-18757750

ABSTRACT

The oxazolidinones represent the first new class of antibiotics to enter into clinical usage within the past 30 years, but their binding site and mechanism of action has not been fully characterized. We have determined the crystal structure of the oxazolidinone linezolid bound to the Deinococcus radiodurans 50S ribosomal subunit. Linezolid binds in the A site pocket at the peptidyltransferase center of the ribosome overlapping the aminoacyl moiety of an A-site bound tRNA as well as many clinically important antibiotics. Binding of linezolid stabilizes a distinct conformation of the universally conserved 23S rRNA nucleotide U2585 that would be nonproductive for peptide bond formation. In conjunction with available biochemical data, we present a model whereby oxazolidinones impart their inhibitory effect by perturbing the correct positioning of tRNAs on the ribosome.


Subject(s)
Anti-Bacterial Agents/chemistry , Oxazolidinones/chemistry , Peptidyl Transferases/chemistry , Peptidyl Transferases/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomes/enzymology , Anti-Bacterial Agents/pharmacology , Binding Sites , Deinococcus/drug effects , Deinococcus/enzymology , Models, Molecular , Nucleic Acid Conformation , Oxazolidinones/pharmacology , Protein Binding , Protein Structure, Tertiary , Ribosomes/drug effects
18.
Sci Adv ; 7(23)2021 06.
Article in English | MEDLINE | ID: mdl-34088665

ABSTRACT

While a structural description of the molecular mechanisms guiding ribosome assembly in eukaryotic systems is emerging, bacteria use an unrelated core set of assembly factors for which high-resolution structural information is still missing. To address this, we used single-particle cryo-electron microscopy to visualize the effects of bacterial ribosome assembly factors RimP, RbfA, RsmA, and RsgA on the conformational landscape of the 30S ribosomal subunit and obtained eight snapshots representing late steps in the folding of the decoding center. Analysis of these structures identifies a conserved secondary structure switch in the 16S ribosomal RNA central to decoding site maturation and suggests both a sequential order of action and molecular mechanisms for the assembly factors in coordinating and controlling this switch. Structural and mechanistic parallels between bacterial and eukaryotic systems indicate common folding features inherent to all ribosomes.


Subject(s)
Ribosome Subunits, Small, Bacterial , Ribosomes , Cryoelectron Microscopy , RNA, Ribosomal, 16S/genetics , Ribosome Subunits, Small
19.
Biomol NMR Assign ; 14(2): 189-193, 2020 10.
Article in English | MEDLINE | ID: mdl-32303998

ABSTRACT

Ribosome biogenesis is an energetically expensive and complex cellular process that involves the coordinated folding of the ribosomal RNA and dozens of ribosomal proteins. It proceeds along multiple parallel pathways and is guided by trans-acting factors called ribosome assembly factors. Although this process has been studied for decades, there are still many open questions regarding the role of the ribosome assembly factors in directing the folding of ribosome biogenesis intermediates. RimP is one of the early acting factors and guides the assembly of the small 30S ribosomal subunit by facilitating the binding of ribosomal proteins uS5 and uS12. Here we report the virtually complete 1H, 15N, and 13C chemical shift assignment of RimP from Escherichia coli. The NMR chemical shift data, deposited in the BMRB data bank under Accession No. 28014, indicates a widely folded protein composed of three alpha helices and eight beta strands.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Nuclear Magnetic Resonance, Biomolecular , Ribosomal Proteins/chemistry , Carbon-13 Magnetic Resonance Spectroscopy , Nitrogen Isotopes , Protein Structure, Secondary
20.
Biomol NMR Assign ; 14(2): 317-321, 2020 10.
Article in English | MEDLINE | ID: mdl-32671633

ABSTRACT

RbfA (ribosome binding factor A; 15.2 kDa) is a protein involved in ribosome biogenesis and has been shown to be important for growth at low temperatures and to act as a suppressor for a cold-sensitive mutation (C23U) in the ribosomal RNA of the small 30S ribosomal subunit. The 3D structure of isolated RbfA has been determined from several organisms showing that RbfA has type-II KH-domain fold topology similar to the KH domain of another assembly factor, Era, whose overexpression can compensate for the deletion of rbfA, suppressing both the cold sensitivity and abnormal accumulation of 17S rRNA in rbfA knockout stains. Interestingly, a RbfAΔ25 variant used in previous NMR studies, truncated at the C-terminal domain to remove 25 unstructured residues causing aggregation at room temperature, was biologically active in the sense that it could complement a knock-out of wildtype RbfA, although it did not act as a suppressor for a 16S cold-sensitive mutation (C23U), nor did it interact stably with the 30S subunit. To complement this work, we report the 1H, 13C, and 15 N backbone and sidechain NMR resonance assignments of full length RbfA from Escherichia coli measured under physiological conditions (pH 7.6). This construct contains seven additional C-terminal residues from the cloning (i.e. one alanine and six residues from the HRV 3C cleavage site) and no aggregation issues were observed over a 1-week period at 293 K. The assignment data has been deposited in the BMRB data bank under Accession No. 27857.


Subject(s)
Escherichia coli Proteins/analysis , Escherichia coli/metabolism , Nuclear Magnetic Resonance, Biomolecular , Ribosomal Proteins/analysis , Ribosomes/metabolism , Amino Acid Sequence , Escherichia coli Proteins/chemistry , Protein Structure, Secondary , Ribosomal Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL