Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746087

ABSTRACT

Eukaryotic ribosome assembly is an intricate process that involves four ribosomal RNAs, 80 ribosomal proteins, and over 200 biogenesis factors that take part in numerous interdependent steps. This complexity creates a large genetic space in which pathogenic mutations can occur. Dead-end ribosome intermediates that result from biogenesis errors are rapidly degraded, affirming the existence of quality control pathway(s) that monitor ribosome assembly. However, the factors that differentiate between on-path and dead-end intermediates are unknown. We engineered a system to perturb ribosome assembly in human cells and discovered that faulty ribosomes are degraded via the ubiquitin proteasome system. We identified ZNF574 as a key component of a novel quality control pathway, which we term the Ribosome Assembly Surveillance Pathway (RASP). Loss of ZNF574 results in the accumulation of faulty biogenesis intermediates that interfere with global ribosome production, further emphasizing the role of RASP in protein homeostasis and cellular health.

2.
Cell Rep Methods ; 4(4): 100755, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38608690

ABSTRACT

In vitro translation is an important method for studying fundamental aspects of co- and post-translational gene regulation, as well as for protein expression in the laboratory and on an industrial scale. Here, by re-examining and improving a human in vitro translation system (HITS), we were able to develop a minimal system where only four components are needed to supplement human cell lysates. Functional characterization of our improved HITS revealed the synergistic effect of mRNA capping and polyadenylation. Furthermore, we found that mRNAs are translated with an efficiency equal to or higher than existing state-of-the-art mammalian in vitro translation systems. Lastly, we present an easy preparation procedure for cytoplasmic extracts from cultured HeLa cells, which can be performed in any cell culture laboratory. These methodological advances will allow HITSs to become a widespread tool in basic molecular biology research.


Subject(s)
Protein Biosynthesis , RNA, Messenger , Humans , HeLa Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Polyadenylation , RNA Caps/metabolism , RNA Caps/genetics
3.
EMBO J ; 43(2): 151-167, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200146

ABSTRACT

Coronaviruses are a group of related RNA viruses that cause respiratory diseases in humans and animals. Understanding the mechanisms of translation regulation during coronaviral infections is critical for developing antiviral therapies and preventing viral spread. Translation of the viral single-stranded RNA genome in the host cell cytoplasm is an essential step in the life cycle of coronaviruses, which affects the cellular mRNA translation landscape in many ways. Here we discuss various viral strategies of translation control, including how members of the Betacoronavirus genus shut down host cell translation and suppress host innate immune functions, as well as the role of the viral non-structural protein 1 (Nsp1) in the process. We also outline the fate of viral RNA, considering stress response mechanisms triggered in infected cells, and describe how unique viral RNA features contribute to programmed ribosomal -1 frameshifting, RNA editing, and translation shutdown evasion.


Subject(s)
Coronavirus Infections , Coronavirus , Animals , Humans , Coronavirus/genetics , Coronavirus Infections/genetics , Betacoronavirus/physiology , Antiviral Agents/pharmacology , RNA, Viral/genetics
4.
Mol Cell ; 83(19): 3546-3557.e8, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37802027

ABSTRACT

Nonstructural protein 1 (Nsp1) produced by coronaviruses inhibits host protein synthesis. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Nsp1 C-terminal domain was shown to bind the ribosomal mRNA channel to inhibit translation, but it is unclear whether this mechanism is broadly used by coronaviruses, whether the Nsp1 N-terminal domain binds the ribosome, or how Nsp1 allows viral RNAs to be translated. Here, we investigated Nsp1 from SARS-CoV-2, Middle East respiratory syndrome coronavirus (MERS-CoV), and Bat-Hp-CoV coronaviruses using structural, biophysical, and biochemical experiments, revealing a conserved role for the C-terminal domain. Additionally, the N-terminal domain of Bat-Hp-CoV Nsp1 binds to the decoding center of the 40S subunit, where it would prevent mRNA and eIF1A accommodation. Structure-based experiments demonstrated the importance of decoding center interactions in all three coronaviruses and showed that the same regions of Nsp1 are necessary for the selective translation of viral RNAs. Our results provide a mechanistic framework to understand how Nsp1 controls preferential translation of viral RNAs.


Subject(s)
COVID-19 , Chiroptera , Animals , Chiroptera/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Protein Domains , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
5.
bioRxiv ; 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37398176

ABSTRACT

Nonstructural protein 1 (Nsp1) produced by coronaviruses shuts down host protein synthesis in infected cells. The C-terminal domain of SARS-CoV-2 Nsp1 was shown to bind to the small ribosomal subunit to inhibit translation, but it is not clear whether this mechanism is broadly used by coronaviruses, whether the N-terminal domain of Nsp1 binds the ribosome, or how Nsp1 specifically permits translation of viral mRNAs. Here, we investigated Nsp1 from three representative Betacoronaviruses - SARS-CoV-2, MERS-CoV, and Bat-Hp-CoV - using structural, biophysical, and biochemical assays. We revealed a conserved mechanism of host translational shutdown across the three coronaviruses. We further demonstrated that the N-terminal domain of Bat-Hp-CoV Nsp1 binds to the decoding center of the 40S subunit, where it would prevent mRNA and eIF1A binding. Structure-based biochemical experiments identified a conserved role of these inhibitory interactions in all three coronaviruses and showed that the same regions of Nsp1 are responsible for the preferential translation of viral mRNAs. Our results provide a mechanistic framework to understand how Betacoronaviruses overcome translational inhibition to produce viral proteins.

6.
Nat Chem ; 15(10): 1431-1443, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37400597

ABSTRACT

DNA-encoded chemical libraries (DELs) consist of large chemical compound collections individually linked to DNA barcodes, facilitating pooled construction and screening. However, screening campaigns often fail if the molecular arrangement of the building blocks is not conducive to an efficient interaction with a protein target. Here we postulated that the use of rigid, compact and stereo-defined central scaffolds for DEL synthesis may facilitate the discovery of very specific ligands capable of discriminating between closely related protein targets. We synthesized a DEL comprising 3,735,936 members, featuring the four stereoisomers of 4-aminopyrrolidine-2-carboxylic acid as central scaffolds. The library was screened in comparative selections against pharmaceutically relevant targets and their closely related protein isoforms. Hit validation results revealed a strong impact of stereochemistry, with large affinity differences between stereoisomers. We identified potent isozyme-selective ligands against multiple protein targets. Some of these hits, specific to tumour-associated antigens, demonstrated tumour-selective targeting in vitro and in vivo. Collectively, constructing DELs with stereo-defined elements contributed to high library productivity and ligand selectivity.

7.
Science ; 380(6651): 1238-1243, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37347872

ABSTRACT

N-terminal methionine excision from newly synthesized proteins, catalyzed cotranslationally by methionine aminopeptidases (METAPs), is an essential and universally conserved process that plays a key role in cell homeostasis and protein biogenesis. However, how METAPs interact with ribosomes and how their cleavage specificity is ensured is unknown. We discovered that in eukaryotes the nascent polypeptide-associated complex (NAC) controls ribosome binding of METAP1. NAC recruits METAP1 using a long, flexible tail and provides a platform for the formation of an active methionine excision complex at the ribosomal tunnel exit. This mode of interaction ensures the efficient excision of methionine from cytosolic proteins, whereas proteins targeted to the endoplasmic reticulum are spared. Our results suggest a broader mechanism for how access of protein biogenesis factors to translating ribosomes is controlled.


Subject(s)
Methionine , Methionyl Aminopeptidases , Protein Biosynthesis , Methionine/metabolism , Methionyl Aminopeptidases/metabolism , Ribosomes/metabolism , Humans , Animals
8.
Nat Struct Mol Biol ; 30(6): 770-777, 2023 06.
Article in English | MEDLINE | ID: mdl-37170030

ABSTRACT

The translocon-associated protein (TRAP) complex resides in the endoplasmic reticulum (ER) membrane and interacts with the Sec translocon and the ribosome to facilitate biogenesis of secretory and membrane proteins. TRAP plays a key role in the secretion of many hormones, including insulin. Here we reveal the molecular architecture of the mammalian TRAP complex and how it engages the translating ribosome associated with Sec61 translocon on the ER membrane. The TRAP complex is anchored to the ribosome via a long tether and its position is further stabilized by a finger-like loop. This positions a cradle-like lumenal domain of TRAP below the translocon for interactions with translocated nascent chains. Our structure-guided TRAP mutations in Caenorhabditis elegans lead to growth deficits associated with increased ER stress and defects in protein hormone secretion. These findings elucidate the molecular basis of the TRAP complex in the biogenesis and translocation of proteins at the ER.


Subject(s)
Endoplasmic Reticulum , Membrane Glycoproteins , Animals , Membrane Glycoproteins/metabolism , Endoplasmic Reticulum/metabolism , Calcium-Binding Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , SEC Translocation Channels/metabolism , Protein Transport , Mammals/metabolism
9.
Science ; 380(6644): 531-536, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37141370

ABSTRACT

The genetic code that specifies the identity of amino acids incorporated into proteins during protein synthesis is almost universally conserved. Mitochondrial genomes feature deviations from the standard genetic code, including the reassignment of two arginine codons to stop codons. The protein required for translation termination at these noncanonical stop codons to release the newly synthesized polypeptides is not currently known. In this study, we used gene editing and ribosomal profiling in combination with cryo-electron microscopy to establish that mitochondrial release factor 1 (mtRF1) detects noncanonical stop codons in human mitochondria by a previously unknown mechanism of codon recognition. We discovered that binding of mtRF1 to the decoding center of the ribosome stabilizes a highly unusual conformation in the messenger RNA in which the ribosomal RNA participates in specific recognition of the noncanonical stop codons.


Subject(s)
Codon, Terminator , Mitochondria , Peptide Chain Termination, Translational , Peptide Termination Factors , Humans , Cryoelectron Microscopy , Mitochondria/genetics , Mitochondria/metabolism , Peptide Termination Factors/chemistry , Protein Conformation
10.
iScience ; 26(2): 105928, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36619367

ABSTRACT

Effective public health measures against SARS-CoV-2 require granular knowledge of population-level immune responses. We developed a Tripartite Automated Blood Immunoassay (TRABI) to assess the IgG response against three SARS-CoV-2 proteins. We used TRABI for continuous seromonitoring of hospital patients and blood donors (n = 72'250) in the canton of Zurich from December 2019 to December 2020 (pre-vaccine period). We found that antibodies waned with a half-life of 75 days, whereas the cumulative incidence rose from 2.3% in June 2020 to 12.2% in mid-December 2020. A follow-up health survey indicated that about 10% of patients infected with wildtype SARS-CoV-2 sustained some symptoms at least twelve months post COVID-19. Crucially, we found no evidence of a difference in long-term complications between those whose infection was symptomatic and those with asymptomatic acute infection. The cohort of asymptomatic SARS-CoV-2-infected subjects represents a resource for the study of chronic and possibly unexpected sequelae.

11.
Science ; 375(6583): 839-844, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35201867

ABSTRACT

The nascent polypeptide-associated complex (NAC) interacts with newly synthesized proteins at the ribosomal tunnel exit and competes with the signal recognition particle (SRP) to prevent mistargeting of cytosolic and mitochondrial polypeptides to the endoplasmic reticulum (ER). How NAC antagonizes SRP and how this is overcome by ER targeting signals are unknown. Here, we found that NAC uses two domains with opposing effects to control SRP access. The core globular domain prevented SRP from binding to signal-less ribosomes, whereas a flexibly attached domain transiently captured SRP to permit scanning of nascent chains. The emergence of an ER-targeting signal destabilized NAC's globular domain and facilitated SRP access to the nascent chain. These findings elucidate how NAC hands over the signal sequence to SRP and imparts specificity of protein localization.


Subject(s)
Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Protein Sorting Signals , Signal Recognition Particle/metabolism , Animals , Binding Sites , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Humans , Models, Molecular , Molecular Chaperones/chemistry , Protein Binding , Protein Domains , Protein Transport , Ribosomes/metabolism , Signal Recognition Particle/chemistry , Ubiquitin/metabolism
12.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35042777

ABSTRACT

Mitochondrial ribosomes (mitoribosomes) play a central role in synthesizing mitochondrial inner membrane proteins responsible for oxidative phosphorylation. Although mitoribosomes from different organisms exhibit considerable structural variations, recent insights into mitoribosome assembly suggest that mitoribosome maturation follows common principles and involves a number of conserved assembly factors. To investigate the steps involved in the assembly of the mitoribosomal small subunit (mt-SSU) we determined the cryoelectron microscopy structures of middle and late assembly intermediates of the Trypanosoma brucei mitochondrial small subunit (mt-SSU) at 3.6- and 3.7-Å resolution, respectively. We identified five additional assembly factors that together with the mitochondrial initiation factor 2 (mt-IF-2) specifically interact with functionally important regions of the rRNA, including the decoding center, thereby preventing premature mRNA or large subunit binding. Structural comparison of assembly intermediates with mature mt-SSU combined with RNAi experiments suggests a noncanonical role of mt-IF-2 and a stepwise assembly process, where modular exchange of ribosomal proteins and assembly factors together with mt-IF-2 ensure proper 9S rRNA folding and protein maturation during the final steps of assembly.


Subject(s)
Mitochondrial Proteins/chemistry , Mitochondrial Ribosomes/chemistry , Oxidative Phosphorylation , RNA, Ribosomal/chemistry , Ribosomal Proteins/chemistry , Ribosome Subunits/chemistry , Cell Line , Cryoelectron Microscopy , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Ribosomes/metabolism , Models, Molecular , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosome Subunits/genetics , Ribosome Subunits/metabolism , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism
13.
Protein Sci ; 31(9): e4418, 2022 09.
Article in English | MEDLINE | ID: mdl-36757682

ABSTRACT

Isoleucyl-tRNA synthetase (IleRS) is an essential enzyme that covalently couples isoleucine to the corresponding tRNA. Bacterial IleRSs group in two clades, ileS1 and ileS2, the latter bringing resistance to the natural antibiotic mupirocin. Generally, bacteria rely on either ileS1 or ileS2 as a standalone housekeeping gene. However, we have found an exception by noticing that Bacillus species with genomic ileS2 consistently also keep ileS1, which appears mandatory in the family Bacillaceae. Taking Priestia (Bacillus) megaterium as a model organism, we showed that PmIleRS1 is constitutively expressed, while PmIleRS2 is stress-induced. Both enzymes share the same level of the aminoacylation accuracy. Yet, PmIleRS1 exhibited a two-fold faster aminoacylation turnover (kcat ) than PmIleRS2 and permitted a notably faster cell-free translation. At the same time, PmIleRS2 displayed a 104 -fold increase in its Ki for mupirocin, arguing that the aminoacylation turnover in IleRS2 could have been traded-off for antibiotic resistance. As expected, a P. megaterium strain deleted for ileS2 was mupirocin-sensitive. Interestingly, an attempt to construct a mupirocin-resistant strain lacking ileS1, a solution not found among species of the family Bacillaceae in nature, led to a viable but compromised strain. Our data suggest that PmIleRS1 is kept to promote fast translation, whereas PmIleRS2 is maintained to provide antibiotic resistance when needed. This is consistent with an emerging picture in which fast-growing organisms predominantly use IleRS1 for competitive survival.


Subject(s)
Bacillus , Drug Resistance, Microbial , Isoleucine-tRNA Ligase , Amino Acyl-tRNA Synthetases/genetics , Bacillus/genetics , Bacteria/genetics , Drug Resistance, Microbial/genetics , Isoleucine-tRNA Ligase/genetics , Mupirocin/pharmacology , RNA, Transfer
14.
Sci Adv ; 7(49): eabl4064, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34851662

ABSTRACT

Transcriptional activator PafBC is the key regulator of the mycobacterial DNA damage response and controls around 150 genes, including genes involved in the canonical SOS response, through an unknown molecular mechanism. Using a combination of biochemistry and cryo­electron microscopy, we demonstrate that PafBC in the presence of single-stranded DNA activates transcription by reprogramming the canonical −10 and −35 promoter specificity of RNA polymerase associated with the housekeeping sigma subunit. We determine the structure of this transcription initiation complex, revealing a unique mode of promoter recognition, which we term "sigma adaptation." PafBC inserts between DNA and sigma factor to mediate recognition of hybrid promoters lacking the −35 but featuring the canonical −10 and a PafBC-specific −26 element. Sigma adaptation may constitute a more general mechanism of transcriptional control in mycobacteria.

15.
Nat Commun ; 12(1): 6635, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34789727

ABSTRACT

Pupylation is the post-translational modification of lysine side chains with prokaryotic ubiquitin-like protein (Pup) that targets proteins for proteasomal degradation in mycobacteria and other members of Actinobacteria. Pup ligase PafA and depupylase Dop are the two enzymes acting in this pathway. Although they share close structural and sequence homology indicative of a common evolutionary origin, they catalyze opposing reactions. Here, we report a series of high-resolution crystal structures of Dop in different functional states along the reaction pathway, including Pup-bound states in distinct conformations. In combination with biochemical analysis, the structures explain the role of the C-terminal residue of Pup in ATP hydrolysis, the process that generates the catalytic phosphate in the active site, and suggest a role for the Dop-loop as an allosteric sensor for Pup-binding and ATP cleavage.


Subject(s)
Amidohydrolases/chemistry , Bacterial Proteins/chemistry , Phosphates/chemistry , Ubiquitins/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Amidohydrolases/metabolism , Bacterial Proteins/metabolism , Catalysis , Catalytic Domain , Crystallography, X-Ray , Hydrogen Bonding , Hydrolysis , Models, Molecular , Phosphates/metabolism , Protein Conformation , Protein Processing, Post-Translational , Ubiquitins/metabolism
16.
Cell Rep ; 36(2): 109350, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260909

ABSTRACT

Co-translational protein targeting to membranes by the signal recognition particle (SRP) is a universally conserved pathway from bacteria to humans. In mammals, SRP and its receptor (SR) have many additional RNA features and protein components compared to the bacterial system, which were recently shown to play regulatory roles. Due to its complexity, the mammalian SRP targeting process is mechanistically not well understood. In particular, it is not clear how SRP recognizes translating ribosomes with exposed signal sequences and how the GTPase activity of SRP and SR is regulated. Here, we present electron cryo-microscopy structures of SRP and SRP·SR in complex with the translating ribosome. The structures reveal the specific molecular interactions between SRP and the emerging signal sequence and the elements that regulate GTPase activity of SRP·SR. Our results suggest the molecular mechanism of how eukaryote-specific elements regulate the early and late stages of SRP-dependent protein targeting.


Subject(s)
Mammals/metabolism , Signal Recognition Particle/metabolism , Animals , Bacteria/metabolism , Cryoelectron Microscopy , GTP Phosphohydrolases/metabolism , Humans , Models, Biological , Models, Molecular , Protein Domains , Protein Transport , RNA/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/ultrastructure , Receptors, Peptide/chemistry , Receptors, Peptide/metabolism , Receptors, Peptide/ultrastructure , Signal Recognition Particle/chemistry , Signal Recognition Particle/ultrastructure
17.
Nucleic Acids Res ; 49(11): 6389-6398, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34086932

ABSTRACT

Biogenesis of ribosomal subunits involves enzymatic modifications of rRNA that fine-tune functionally important regions. The universally conserved prokaryotic dimethyltransferase KsgA sequentially modifies two universally conserved adenosine residues in helix 45 of the small ribosomal subunit rRNA, which is in proximity of the decoding site. Here we present the cryo-EM structure of Escherichia coli KsgA bound to an E. coli 30S at a resolution of 3.1 Å. The high-resolution structure reveals how KsgA recognizes immature rRNA and binds helix 45 in a conformation where one of the substrate nucleotides is flipped-out into the active site. We suggest that successive processing of two adjacent nucleotides involves base-flipping of the rRNA, which allows modification of the second substrate nucleotide without dissociation of the enzyme. Since KsgA is homologous to the essential eukaryotic methyltransferase Dim1 involved in 40S maturation, these results have also implications for understanding eukaryotic ribosome maturation.


Subject(s)
Adenosine/metabolism , Escherichia coli/enzymology , Methyltransferases/chemistry , Adenosine/chemistry , Cryoelectron Microscopy , Methyltransferases/metabolism , Models, Molecular , Protein Conformation , Ribosome Subunits, Small, Bacterial/chemistry , Substrate Specificity
18.
Nat Commun ; 12(1): 3671, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135320

ABSTRACT

Mitochondrial ribosomes are specialized for the synthesis of membrane proteins responsible for oxidative phosphorylation. Mammalian mitoribosomes have diverged considerably from the ancestral bacterial ribosomes and feature dramatically reduced ribosomal RNAs. The structural basis of the mammalian mitochondrial ribosome assembly is currently not well understood. Here we present eight distinct assembly intermediates of the human large mitoribosomal subunit involving seven assembly factors. We discover that the NSUN4-MTERF4 dimer plays a critical role in the process by stabilizing the 16S rRNA in a conformation that exposes the functionally important regions of rRNA for modification by the MRM2 methyltransferase and quality control interactions with the conserved mitochondrial GTPase MTG2 that contacts the sarcin-ricin loop and the immature active site. The successive action of these factors leads to the formation of the peptidyl transferase active site of the mitoribosome and the folding of the surrounding rRNA regions responsible for interactions with tRNAs and the small ribosomal subunit.


Subject(s)
Mitochondrial Ribosomes/chemistry , Peptidyl Transferases/chemistry , Catalytic Domain , Cryoelectron Microscopy , Humans , Methyltransferases/chemistry , Methyltransferases/metabolism , Mitochondrial Ribosomes/metabolism , Models, Molecular , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/metabolism , Nucleic Acid Conformation , Peptidyl Transferases/metabolism , Protein Multimerization , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , Ribosome Subunits, Large/chemistry , Ribosome Subunits, Large/metabolism , Transcription Factors/metabolism
19.
Science ; 372(6547): 1220-1224, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34112695

ABSTRACT

Viruses are ubiquitous pathogens of global impact. Prompted by the hypothesis that their earliest progenitors recruited host proteins for virion formation, we have used stringent laboratory evolution to convert a bacterial enzyme that lacks affinity for nucleic acids into an artificial nucleocapsid that efficiently packages and protects multiple copies of its own encoding messenger RNA. Revealing remarkable convergence on the molecular hallmarks of natural viruses, the accompanying changes reorganized the protein building blocks into an interlaced 240-subunit icosahedral capsid that is impermeable to nucleases, and emergence of a robust RNA stem-loop packaging cassette ensured high encapsidation yields and specificity. In addition to evincing a plausible evolutionary pathway for primordial viruses, these findings highlight practical strategies for developing nonviral carriers for diverse vaccine and delivery applications.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Capsid/metabolism , Directed Molecular Evolution , RNA, Messenger/metabolism , Amino Acid Substitution , Aquifex/enzymology , Bacterial Proteins/chemistry , Capsid/chemistry , Cryoelectron Microscopy , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Nucleocapsid/chemistry , Nucleocapsid/genetics , Nucleocapsid/metabolism , Protein Domains , Protein Structure, Secondary , Protein Subunits , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribonucleases/metabolism
20.
Sci Adv ; 7(21)2021 05.
Article in English | MEDLINE | ID: mdl-34020957

ABSTRACT

The conserved signal recognition particle (SRP) cotranslationally delivers ~30% of the proteome to the eukaryotic endoplasmic reticulum (ER). The molecular mechanism by which eukaryotic SRP transitions from cargo recognition in the cytosol to protein translocation at the ER is not understood. Here, structural, biochemical, and single-molecule studies show that this transition requires multiple sequential conformational rearrangements in the targeting complex initiated by guanosine triphosphatase (GTPase)-driven compaction of the SRP receptor (SR). Disruption of these rearrangements, particularly in mutant SRP54G226E linked to severe congenital neutropenia, uncouples the SRP/SR GTPase cycle from protein translocation. Structures of targeting intermediates reveal the molecular basis of early SRP-SR recognition and emphasize the role of eukaryote-specific elements in regulating targeting. Our results provide a molecular model for the structural and functional transitions of SRP throughout the targeting cycle and show that these transitions provide important points for biological regulation that can be perturbed in genetic diseases.

SELECTION OF CITATIONS
SEARCH DETAIL
...