Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 182(6): 1508-1518.e16, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32783917

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most frequent cause of familial Parkinson's disease. LRRK2 is a multi-domain protein containing a kinase and GTPase. Using correlative light and electron microscopy, in situ cryo-electron tomography, and subtomogram analysis, we reveal a 14-Å structure of LRRK2 bearing a pathogenic mutation that oligomerizes as a right-handed double helix around microtubules, which are left-handed. Using integrative modeling, we determine the architecture of LRRK2, showing that the GTPase and kinase are in close proximity, with the GTPase closer to the microtubule surface, whereas the kinase is exposed to the cytoplasm. We identify two oligomerization interfaces mediated by non-catalytic domains. Mutation of one of these abolishes LRRK2 microtubule-association. Our work demonstrates the power of cryo-electron tomography to generate models of previously unsolved structures in their cellular environment.


Subject(s)
Cryoelectron Microscopy/methods , Electron Microscope Tomography/methods , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Microtubules/metabolism , Parkinson Disease/metabolism , Cytoplasm/metabolism , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , HEK293 Cells , Humans , Microscopy, Electron, Transmission , Microtubules/chemistry , Models, Chemical , Mutation , Parkinson Disease/genetics , Parkinson Disease/pathology , Phosphotransferases/chemistry , Phosphotransferases/metabolism , Protein Domains , WD40 Repeats
2.
Mol Cell ; 83(4): 607-621.e4, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36804914

ABSTRACT

Ribosome-associated quality control (RQC) is a conserved process degrading potentially toxic truncated nascent peptides whose malfunction underlies neurodegeneration and proteostasis decline in aging. During RQC, dissociation of stalled ribosomes is followed by elongation of the nascent peptide with alanine and threonine residues, driven by Rqc2 independently of mRNA, the small ribosomal subunit and guanosine triphosphate (GTP)-hydrolyzing factors. The resulting CAT tails (carboxy-terminal tails) and ubiquitination by Ltn1 mark nascent peptides for proteasomal degradation. Here we present ten cryogenic electron microscopy (cryo-EM) structures, revealing the mechanistic basis of individual steps of the CAT tailing cycle covering initiation, decoding, peptidyl transfer, and tRNA translocation. We discovered eIF5A as a crucial eukaryotic RQC factor enabling peptidyl transfer. Moreover, we observed dynamic behavior of RQC factors and tRNAs allowing for processivity of the CAT tailing cycle without additional energy input. Together, these results elucidate key differences as well as common principles between CAT tailing and canonical translation.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Protein Biosynthesis , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Peptides/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Quality Control
3.
Mol Cell ; 82(18): 3424-3437.e8, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36113412

ABSTRACT

Cells can respond to stalled ribosomes by sensing ribosome collisions and employing quality control pathways. How ribosome stalling is resolved without collisions, however, has remained elusive. Here, focusing on noncolliding stalling exhibited by decoding-defective ribosomes, we identified Fap1 as a stalling sensor triggering 18S nonfunctional rRNA decay via polyubiquitination of uS3. Ribosome profiling revealed an enrichment of Fap1 at the translation initiation site but also an association with elongating individual ribosomes. Cryo-EM structures of Fap1-bound ribosomes elucidated Fap1 probing the mRNA simultaneously at both the entry and exit channels suggesting an mRNA stasis sensing activity, and Fap1 sterically hinders the formation of canonical collided di-ribosomes. Our findings indicate that individual stalled ribosomes are the potential signal for ribosome dysfunction, leading to accelerated turnover of the ribosome itself.


Subject(s)
Protein Biosynthesis , Ribosomes , RNA Stability , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Ribosomes/metabolism
4.
Nature ; 603(7901): 503-508, 2022 03.
Article in English | MEDLINE | ID: mdl-35264790

ABSTRACT

Ribosome rescue pathways recycle stalled ribosomes and target problematic mRNAs and aborted proteins for degradation1,2. In bacteria, it remains unclear how rescue pathways distinguish ribosomes stalled in the middle of a transcript from actively translating ribosomes3-6. Here, using a genetic screen in Escherichia coli, we discovered a new rescue factor that has endonuclease activity. SmrB cleaves mRNAs upstream of stalled ribosomes, allowing the ribosome rescue factor tmRNA (which acts on truncated mRNAs3) to rescue upstream ribosomes. SmrB is recruited to ribosomes and is activated by collisions. Cryo-electron microscopy structures of collided disomes from E. coli and Bacillus subtilis show distinct and conserved arrangements of individual ribosomes and the composite SmrB-binding site. These findings reveal the underlying mechanisms by which ribosome collisions trigger ribosome rescue in bacteria.


Subject(s)
Escherichia coli , Ribosomes , Bacteria/genetics , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Biosynthesis , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism
5.
EMBO J ; 40(11): e102277, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33876849

ABSTRACT

The ongoing outbreak of severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) demonstrates the continuous threat of emerging coronaviruses (CoVs) to public health. SARS-CoV-2 and SARS-CoV share an otherwise non-conserved part of non-structural protein 3 (Nsp3), therefore named as "SARS-unique domain" (SUD). We previously found a yeast-2-hybrid screen interaction of the SARS-CoV SUD with human poly(A)-binding protein (PABP)-interacting protein 1 (Paip1), a stimulator of protein translation. Here, we validate SARS-CoV SUD:Paip1 interaction by size-exclusion chromatography, split-yellow fluorescent protein, and co-immunoprecipitation assays, and confirm such interaction also between the corresponding domain of SARS-CoV-2 and Paip1. The three-dimensional structure of the N-terminal domain of SARS-CoV SUD ("macrodomain II", Mac2) in complex with the middle domain of Paip1, determined by X-ray crystallography and small-angle X-ray scattering, provides insights into the structural determinants of the complex formation. In cellulo, SUD enhances synthesis of viral but not host proteins via binding to Paip1 in pBAC-SARS-CoV replicon-transfected cells. We propose a possible mechanism for stimulation of viral translation by the SUD of SARS-CoV and SARS-CoV-2.


Subject(s)
Coronavirus Papain-Like Proteases/metabolism , Gene Expression Regulation, Viral , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins , Chromatography, Gel , Coronavirus Papain-Like Proteases/chemistry , Crystallography, X-Ray , Genes, Reporter , HEK293 Cells , Humans , Immunoprecipitation , Luminescent Proteins , Models, Molecular , Peptide Initiation Factors/chemistry , Protein Binding , Protein Biosynthesis , Protein Conformation , Protein Domains , Protein Interaction Mapping , RNA, Viral/genetics , RNA-Binding Proteins/chemistry , RNA-Dependent RNA Polymerase/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Ribosome Subunits/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2/genetics , Scattering, Small Angle , Sequence Alignment , Sequence Homology, Amino Acid , Viral Nonstructural Proteins/chemistry , X-Ray Diffraction
6.
EMBO J ; 39(3): e103365, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31858614

ABSTRACT

Inhibitory codon pairs and poly(A) tracts within the translated mRNA cause ribosome stalling and reduce protein output. The molecular mechanisms that drive these stalling events, however, are still unknown. Here, we use a combination of in vitro biochemistry, ribosome profiling, and cryo-EM to define molecular mechanisms that lead to these ribosome stalls. First, we use an in vitro reconstituted yeast translation system to demonstrate that inhibitory codon pairs slow elongation rates which are partially rescued by increased tRNA concentration or by an artificial tRNA not dependent on wobble base-pairing. Ribosome profiling data extend these observations by revealing that paused ribosomes with empty A sites are enriched on these sequences. Cryo-EM structures of stalled ribosomes provide a structural explanation for the observed effects by showing decoding-incompatible conformations of mRNA in the A sites of all studied stall- and collision-inducing sequences. Interestingly, in the case of poly(A) tracts, the inhibitory conformation of the mRNA in the A site involves a nucleotide stacking array. Together, these data demonstrate a novel mRNA-induced mechanisms of translational stalling in eukaryotic ribosomes.


Subject(s)
Protein Biosynthesis , RNA, Messenger/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Codon , Cryoelectron Microscopy , Models, Molecular , Nucleic Acid Conformation , Poly A/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Saccharomyces cerevisiae/genetics
7.
PLoS Biol ; 18(7): e3000780, 2020 07.
Article in English | MEDLINE | ID: mdl-32687489

ABSTRACT

Cells adjust to nutrient deprivation by reversible translational shutdown. This is accompanied by maintaining inactive ribosomes in a hibernation state, in which they are bound by proteins with inhibitory and protective functions. In eukaryotes, such a function was attributed to suppressor of target of Myb protein 1 (Stm1; SERPINE1 mRNA-binding protein 1 [SERBP1] in mammals), and recently, late-annotated short open reading frame 2 (Lso2; coiled-coil domain containing short open reading frame 124 [CCDC124] in mammals) was found to be involved in translational recovery after starvation from stationary phase. Here, we present cryo-electron microscopy (cryo-EM) structures of translationally inactive yeast and human ribosomes. We found Lso2/CCDC124 accumulating on idle ribosomes in the nonrotated state, in contrast to Stm1/SERBP1-bound ribosomes, which display a rotated state. Lso2/CCDC124 bridges the decoding sites of the small with the GTPase activating center (GAC) of the large subunit. This position allows accommodation of the duplication of multilocus region 34 protein (Dom34)-dependent ribosome recycling system, which splits Lso2-containing, but not Stm1-containing, ribosomes. We propose a model in which Lso2 facilitates rapid translation reactivation by stabilizing the recycling-competent state of inactive ribosomes.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Binding Sites , Conserved Sequence , Evolution, Molecular , HEK293 Cells , Humans , Models, Molecular , Peptides/chemistry , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/metabolism , RNA-Binding Proteins/metabolism , Ribosomes/ultrastructure , Saccharomyces cerevisiae/ultrastructure , Structure-Activity Relationship
8.
Nucleic Acids Res ; 49(16): 9539-9547, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34403461

ABSTRACT

In Escherichia coli, elevated levels of free l-tryptophan (l-Trp) promote translational arrest of the TnaC peptide by inhibiting its termination. However, the mechanism by which translation-termination by the UGA-specific decoding release factor 2 (RF2) is inhibited at the UGA stop codon of stalled TnaC-ribosome-nascent chain complexes has so far been ambiguous. This study presents cryo-EM structures for ribosomes stalled by TnaC in the absence and presence of RF2 at average resolutions of 2.9 and 3.5 Å, respectively. Stalled TnaC assumes a distinct conformation composed of two small α-helices that act together with residues in the peptide exit tunnel (PET) to coordinate a single L-Trp molecule. In addition, while the peptidyl-transferase center (PTC) is locked in a conformation that allows RF2 to adopt its canonical position in the ribosome, it prevents the conserved and catalytically essential GGQ motif of RF2 from adopting its active conformation in the PTC. This explains how translation of the TnaC peptide effectively allows the ribosome to function as a L-Trp-specific small-molecule sensor that regulates the tnaCAB operon.


Subject(s)
Escherichia coli Proteins/ultrastructure , Peptide Termination Factors/ultrastructure , Protein Biosynthesis , Ribosomes/ultrastructure , Codon, Terminator/genetics , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli/ultrastructure , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Protein Conformation , Protein Conformation, alpha-Helical , Ribosomes/genetics , Tryptophan/genetics
9.
Nat Chem Biol ; 16(10): 1071-1077, 2020 10.
Article in English | MEDLINE | ID: mdl-32601485

ABSTRACT

The increase in multi-drug resistant pathogenic bacteria is making our current arsenal of clinically used antibiotics obsolete, highlighting the urgent need for new lead compounds with distinct target binding sites to avoid cross-resistance. Here we report that the aromatic polyketide antibiotic tetracenomycin (TcmX) is a potent inhibitor of protein synthesis, and does not induce DNA damage as previously thought. Despite the structural similarity to the well-known translation inhibitor tetracycline, we show that TcmX does not interact with the small ribosomal subunit, but rather binds to the large subunit, within the polypeptide exit tunnel. This previously unappreciated binding site is located adjacent to the macrolide-binding site, where TcmX stacks on the noncanonical basepair formed by U1782 and U2586 of the 23S ribosomal RNA. Although the binding site is distinct from the macrolide antibiotics, our results indicate that like macrolides, TcmX allows translation of short oligopeptides before further translation is blocked.


Subject(s)
Amycolatopsis/drug effects , Gene Expression Regulation, Bacterial/drug effects , Amycolatopsis/genetics , Amycolatopsis/metabolism , Binding Sites , Cryoelectron Microscopy , Drug Resistance, Bacterial , Escherichia coli , HEK293 Cells , Humans , Microbial Sensitivity Tests , Models, Molecular , Mutation , Naphthacenes/chemistry , Naphthacenes/pharmacology , Protein Binding , Protein Biosynthesis/drug effects , Protein Conformation , Ribosomes/metabolism
10.
Nat Commun ; 15(1): 5481, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38942792

ABSTRACT

Tigecycline is widely used for treating complicated bacterial infections for which there are no effective drugs. It inhibits bacterial protein translation by blocking the ribosomal A-site. However, even though it is also cytotoxic for human cells, the molecular mechanism of its inhibition remains unclear. Here, we present cryo-EM structures of tigecycline-bound human mitochondrial 55S, 39S, cytoplasmic 80S and yeast cytoplasmic 80S ribosomes. We find that at clinically relevant concentrations, tigecycline effectively targets human 55S mitoribosomes, potentially, by hindering A-site tRNA accommodation and by blocking the peptidyl transfer center. In contrast, tigecycline does not bind to human 80S ribosomes under physiological concentrations. However, at high tigecycline concentrations, in addition to blocking the A-site, both human and yeast 80S ribosomes bind tigecycline at another conserved binding site restricting the movement of the L1 stalk. In conclusion, the observed distinct binding properties of tigecycline may guide new pathways for drug design and therapy.


Subject(s)
Cryoelectron Microscopy , Ribosomes , Tigecycline , Tigecycline/pharmacology , Tigecycline/chemistry , Humans , Ribosomes/metabolism , Ribosomes/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Binding Sites , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Protein Biosynthesis/drug effects , Mitochondrial Ribosomes/metabolism , Mitochondrial Ribosomes/chemistry , Mitochondrial Ribosomes/drug effects , Models, Molecular , RNA, Transfer/metabolism , RNA, Transfer/chemistry
11.
Elife ; 122023 03 17.
Article in English | MEDLINE | ID: mdl-36929751

ABSTRACT

Biogenesis intermediates of nucleolar ribosomal 60S precursor particles undergo a number of structural maturation steps before they transit to the nucleoplasm and are finally exported into the cytoplasm. The AAA+-ATPase Rea1 participates in the nucleolar exit by releasing the Ytm1-Erb1 heterodimer from the evolving pre-60S particle. Here, we show that the DEAD-box RNA helicase Spb4 with its interacting partner Rrp17 is further integrated into this maturation event. Spb4 binds to a specific class of late nucleolar pre-60S intermediates, whose cryo-EM structure revealed how its helicase activity facilitates melting and restructuring of 25S rRNA helices H62 and H63/H63a prior to Ytm1-Erb1 release. In vitro maturation of such Spb4-enriched pre-60S particles, incubated with purified Rea1 and its associated pentameric Rix1-complex in the presence of ATP, combined with cryo-EM analysis depicted the details of the Rea1-dependent large-scale pre-ribosomal remodeling. Our structural insights unveil how the Rea1 ATPase and Spb4 helicase remodel late nucleolar pre-60S particles by rRNA restructuring and dismantling of a network of several ribosomal assembly factors.


Subject(s)
Adenosine Triphosphatases , Saccharomyces cerevisiae Proteins , Adenosine Triphosphatases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , RNA Helicases/metabolism , RNA, Ribosomal/metabolism , Ribosomes/metabolism , RNA Precursors/metabolism , Ribosomal Proteins/genetics
12.
Nat Commun ; 14(1): 2730, 2023 05 12.
Article in English | MEDLINE | ID: mdl-37169754

ABSTRACT

In actively translating 80S ribosomes the ribosomal protein eS7 of the 40S subunit is monoubiquitinated by the E3 ligase Not4 and deubiquitinated by Otu2 upon ribosomal subunit recycling. Despite its importance for translation efficiency the exact role and structural basis for this translational reset is poorly understood. Here, structural analysis by cryo-electron microscopy of native and reconstituted Otu2-bound ribosomal complexes reveals that Otu2 engages 40S subunits mainly between ribosome recycling and initiation stages. Otu2 binds to several sites on the intersubunit surface of the 40S that are not occupied by any other 40S-binding factors. This binding mode explains the discrimination against 80S ribosomes via the largely helical N-terminal domain of Otu2 as well as the specificity for mono-ubiquitinated eS7 on 40S. Collectively, this study reveals mechanistic insights into the Otu2-driven deubiquitination steps for translational reset during ribosome recycling/(re)initiation.


Subject(s)
Ribosomal Proteins , Ribosomes , Cryoelectron Microscopy , Protein Biosynthesis , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosomes/metabolism
13.
Nat Struct Mol Biol ; 30(8): 1119-1131, 2023 08.
Article in English | MEDLINE | ID: mdl-37291423

ABSTRACT

The 5S ribonucleoprotein (RNP) is assembled from its three components (5S rRNA, Rpl5/uL18 and Rpl11/uL5) before being incorporated into the pre-60S subunit. However, when ribosome synthesis is disturbed, a free 5S RNP can enter the MDM2-p53 pathway to regulate cell cycle and apoptotic signaling. Here we reconstitute and determine the cryo-electron microscopy structure of the conserved hexameric 5S RNP with fungal or human factors. This reveals how the nascent 5S rRNA associates with the initial nuclear import complex Syo1-uL18-uL5 and, upon further recruitment of the nucleolar factors Rpf2 and Rrs1, develops into the 5S RNP precursor that can assemble into the pre-ribosome. In addition, we elucidate the structure of another 5S RNP intermediate, carrying the human ubiquitin ligase Mdm2, which unravels how this enzyme can be sequestered from its target substrate p53. Our data provide molecular insight into how the 5S RNP can mediate between ribosome biogenesis and cell proliferation.


Subject(s)
RNA, Ribosomal, 5S , Tumor Suppressor Protein p53 , Humans , RNA, Ribosomal, 5S/chemistry , Tumor Suppressor Protein p53/metabolism , Cryoelectron Microscopy , Ribosomal Proteins/metabolism , Ribonucleoproteins/metabolism , Ribosomes/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism
14.
Nat Struct Mol Biol ; 27(4): 323-332, 2020 04.
Article in English | MEDLINE | ID: mdl-32203490

ABSTRACT

Ribosome-associated quality control (RQC) represents a rescue pathway in eukaryotic cells that is triggered upon translational stalling. Collided ribosomes are recognized for subsequent dissociation followed by degradation of nascent peptides. However, endogenous RQC-inducing sequences and the mechanism underlying the ubiquitin-dependent ribosome dissociation remain poorly understood. Here, we identified SDD1 messenger RNA from Saccharomyces cerevisiae as an endogenous RQC substrate and reveal the mechanism of its mRNA-dependent and nascent peptide-dependent translational stalling. In vitro translation of SDD1 mRNA enabled the reconstitution of Hel2-dependent polyubiquitination of collided disomes and, preferentially, trisomes. The distinct trisome architecture, visualized using cryo-EM, provides the structural basis for the more-efficient recognition by Hel2 compared with that of disomes. Subsequently, the Slh1 helicase subunit of the RQC trigger (RQT) complex preferentially dissociates the first stalled polyubiquitinated ribosome in an ATP-dependent manner. Together, these findings provide fundamental mechanistic insights into RQC and its physiological role in maintaining cellular protein homeostasis.


Subject(s)
Cell Cycle Proteins/ultrastructure , Protein Biosynthesis , Ribosomes/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Serine Endopeptidases/ultrastructure , Ubiquitin-Protein Ligases/ultrastructure , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Peptides/chemistry , Peptides/genetics , RNA, Messenger/genetics , Ribosomes/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
15.
Science ; 368(6488)2020 04 17.
Article in English | MEDLINE | ID: mdl-32299921

ABSTRACT

Control of messenger RNA (mRNA) decay rate is intimately connected to translation elongation, but the spatial coordination of these events is poorly understood. The Ccr4-Not complex initiates mRNA decay through deadenylation and activation of decapping. We used a combination of cryo-electron microscopy, ribosome profiling, and mRNA stability assays to examine the recruitment of Ccr4-Not to the ribosome via specific interaction of the Not5 subunit with the ribosomal E-site in Saccharomyces cerevisiae This interaction occurred when the ribosome lacked accommodated A-site transfer RNA, indicative of low codon optimality. Loss of the interaction resulted in the inability of the mRNA degradation machinery to sense codon optimality. Our findings elucidate a physical link between the Ccr4-Not complex and the ribosome and provide mechanistic insight into the coupling of decoding efficiency with mRNA stability.


Subject(s)
Codon , Peptide Chain Elongation, Translational , RNA Stability , Repressor Proteins/metabolism , Ribonucleases/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Cryoelectron Microscopy , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Peptide Initiation Factors/metabolism , Protein Conformation, alpha-Helical , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Ribonucleases/chemistry , Ribonucleases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Eukaryotic Translation Initiation Factor 5A
16.
Science ; 369(6508): 1249-1255, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32680882

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the current coronavirus disease 2019 (COVID-19) pandemic. A major virulence factor of SARS-CoVs is the nonstructural protein 1 (Nsp1), which suppresses host gene expression by ribosome association. Here, we show that Nsp1 from SARS-CoV-2 binds to the 40S ribosomal subunit, resulting in shutdown of messenger RNA (mRNA) translation both in vitro and in cells. Structural analysis by cryo-electron microscopy of in vitro-reconstituted Nsp1-40S and various native Nsp1-40S and -80S complexes revealed that the Nsp1 C terminus binds to and obstructs the mRNA entry tunnel. Thereby, Nsp1 effectively blocks retinoic acid-inducible gene I-dependent innate immune responses that would otherwise facilitate clearance of the infection. Thus, the structural characterization of the inhibitory mechanism of Nsp1 may aid structure-based drug design against SARS-CoV-2.


Subject(s)
Betacoronavirus/chemistry , Immune Evasion , Immunity, Innate , Protein Biosynthesis , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Betacoronavirus/immunology , Betacoronavirus/metabolism , Betacoronavirus/physiology , Binding Sites , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cryoelectron Microscopy , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Models, Molecular , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , Protein Structure, Secondary , RNA, Messenger/metabolism , Receptors, Immunologic , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism , SARS-CoV-2
17.
Nat Struct Mol Biol ; 26(4): 275-280, 2019 04.
Article in English | MEDLINE | ID: mdl-30911188

ABSTRACT

Messenger RNA (mRNA) homeostasis represents an essential part of gene expression, in which the generation of mRNA by RNA polymerase is counter-balanced by its degradation by nucleases. The conserved 5'-to-3' exoribonuclease Xrn1 has a crucial role in eukaryotic mRNA homeostasis by degrading decapped or cleaved mRNAs post-translationally and, more surprisingly, also co-translationally. Here we report that active Xrn1 can directly and specifically interact with the translation machinery. A cryo-electron microscopy structure of a programmed Saccharomyces cerevisiae 80S ribosome-Xrn1 nuclease complex reveals how the conserved core of Xrn1 enables binding at the mRNA exit site of the ribosome. This interface provides a conduit for channelling of the mRNA from the ribosomal decoding site directly into the active center of the nuclease, thus separating mRNA decoding from degradation by only 17 ± 1 nucleotides. These findings explain how rapid 5'-to-3' mRNA degradation is coupled efficiently to its final round of mRNA translation.


Subject(s)
Exoribonucleases/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Cryoelectron Microscopy , Exoribonucleases/genetics , Exoribonucleases/ultrastructure , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure
18.
Science ; 355(6321): 194-197, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28082593

ABSTRACT

We observed the assembly of a nucleus-like structure in bacteria during viral infection. Using fluorescence microscopy and cryo-electron tomography, we showed that Pseudomonas chlororaphis phage 201φ2-1 assembled a compartment that separated viral DNA from the cytoplasm. The phage compartment was centered by a bipolar tubulin-based spindle, and it segregated phage and bacterial proteins according to function. Proteins involved in DNA replication and transcription localized inside the compartment, whereas proteins involved in translation and nucleotide synthesis localized outside. Later during infection, viral capsids assembled on the cytoplasmic membrane and moved to the surface of the compartment for DNA packaging. Ultimately, viral particles were released from the compartment and the cell lysed. These results demonstrate that phages have evolved a specialized structure to compartmentalize viral replication.


Subject(s)
Pseudomonas Phages/physiology , Pseudomonas chlororaphis/virology , Virus Assembly , Capsid/metabolism , Capsid Proteins/biosynthesis , Capsid Proteins/genetics , Cryoelectron Microscopy , Cytoplasm/ultrastructure , Cytoplasm/virology , DNA, Viral/biosynthesis , Microscopy, Fluorescence , Pseudomonas Phages/genetics , Pseudomonas chlororaphis/ultrastructure , Transcription, Genetic
19.
Cell Rep ; 17(11): 2943-2954, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27974208

ABSTRACT

Members of the YidC/Oxa1/Alb3 family universally facilitate membrane protein biogenesis, via mechanisms that have thus far remained unclear. Here, we investigated two crucial functional aspects: the interaction of YidC with ribosome:nascent chain complexes (RNCs) and the structural dynamics of RNC-bound YidC in nanodiscs. We observed that a fully exposed nascent transmembrane domain (TMD) is required for high-affinity YidC:RNC interactions, while weaker binding may already occur at earlier stages of translation. YidC efficiently catalyzed the membrane insertion of nascent TMDs in both fluid and gel phase membranes. Cryo-electron microscopy and fluorescence analysis revealed a conformational change in YidC upon nascent chain insertion: the essential TMDs 2 and 3 of YidC were tilted, while the amphipathic helix EH1 relocated into the hydrophobic core of the membrane. We suggest that EH1 serves as a mechanical lever, facilitating a coordinated movement of YidC TMDs to trigger the release of nascent chains into the membrane.


Subject(s)
Escherichia coli Proteins/genetics , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Protein Biosynthesis , Ribosomes/metabolism , Cryoelectron Microscopy , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Ribosomes/genetics , Ribosomes/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL