Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 377
Filter
1.
Nat Commun ; 15(1): 3945, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730238

ABSTRACT

Proline-rich antimicrobial peptides (PrAMPs) inhibit bacterial protein biosynthesis by binding to the polypeptide exit tunnel (PET) near the peptidyl transferase center. Api137, an optimized derivative of honeybee PrAMP apidaecin, inhibits protein expression by trapping release factors (RFs), which interact with stop codons on ribosomes to terminate translation. This study uses cryo-EM, functional assays and molecular dynamic (MD) simulations to show that Api137 additionally occupies a second binding site near the exit of the PET and can repress translation independently of RF-trapping. Api88, a C-terminally amidated (-CONH2) analog of Api137 (-COOH), binds to the same sites, occupies a third binding pocket and interferes with the translation process presumably without RF-trapping. In conclusion, apidaecin-derived PrAMPs inhibit bacterial ribosomes by multimodal mechanisms caused by minor structural changes and thus represent a promising pool for drug development efforts.


Subject(s)
Antimicrobial Cationic Peptides , Molecular Dynamics Simulation , Ribosomes , Ribosomes/metabolism , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Protein Biosynthesis , Binding Sites , Cryoelectron Microscopy , Escherichia coli/metabolism , Escherichia coli/genetics , Escherichia coli/drug effects , Peptide Termination Factors/metabolism , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Protein Binding , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/pharmacology
2.
Nature ; 626(8001): 1125-1132, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355796

ABSTRACT

To conserve energy during starvation and stress, many organisms use hibernation factor proteins to inhibit protein synthesis and protect their ribosomes from damage1,2. In bacteria, two families of hibernation factors have been described, but the low conservation of these proteins and the huge diversity of species, habitats and environmental stressors have confounded their discovery3-6. Here, by combining cryogenic electron microscopy, genetics and biochemistry, we identify Balon, a new hibernation factor in the cold-adapted bacterium Psychrobacter urativorans. We show that Balon is a distant homologue of the archaeo-eukaryotic translation factor aeRF1 and is found in 20% of representative bacteria. During cold shock or stationary phase, Balon occupies the ribosomal A site in both vacant and actively translating ribosomes in complex with EF-Tu, highlighting an unexpected role for EF-Tu in the cellular stress response. Unlike typical A-site substrates, Balon binds to ribosomes in an mRNA-independent manner, initiating a new mode of ribosome hibernation that can commence while ribosomes are still engaged in protein synthesis. Our work suggests that Balon-EF-Tu-regulated ribosome hibernation is a ubiquitous bacterial stress-response mechanism, and we demonstrate that putative Balon homologues in Mycobacteria bind to ribosomes in a similar fashion. This finding calls for a revision of the current model of ribosome hibernation inferred from common model organisms and holds numerous implications for how we understand and study ribosome hibernation.


Subject(s)
Bacterial Proteins , Cold-Shock Response , Peptide Termination Factors , Protein Biosynthesis , Psychrobacter , Ribosomal Proteins , Ribosomes , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factor Tu/ultrastructure , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/ultrastructure , Ribosomes/chemistry , Ribosomes/metabolism , Ribosomes/ultrastructure , Psychrobacter/chemistry , Psychrobacter/genetics , Psychrobacter/metabolism , Psychrobacter/ultrastructure , Cryoelectron Microscopy , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Peptide Termination Factors/ultrastructure
3.
Nat Chem Biol ; 20(5): 605-614, 2024 May.
Article in English | MEDLINE | ID: mdl-38267667

ABSTRACT

In response to environmental changes, cells flexibly and rapidly alter gene expression through translational controls. In plants, the translation of NIP5;1, a boric acid diffusion facilitator, is downregulated in response to an excess amount of boric acid in the environment through upstream open reading frames (uORFs) that consist of only AUG and stop codons. However, the molecular details of how this minimum uORF controls translation of the downstream main ORF in a boric acid-dependent manner have remained unclear. Here, by combining ribosome profiling, translation complex profile sequencing, structural analysis with cryo-electron microscopy and biochemical assays, we show that the 80S ribosome assembled at AUG-stop migrates into the subsequent RNA segment, followed by downstream translation initiation, and that boric acid impedes this process by the stable confinement of eukaryotic release factor 1 on the 80S ribosome on AUG-stop. Our results provide molecular insight into translation regulation by a minimum and environment-responsive uORF.


Subject(s)
Boric Acids , Protein Biosynthesis , Ribosomes , Ribosomes/metabolism , Boric Acids/chemistry , Peptide Termination Factors/metabolism , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Cryoelectron Microscopy , Open Reading Frames , Codon, Terminator , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics
4.
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
5.
Mol Cell Biol ; 43(4): 157-173, 2023.
Article in English | MEDLINE | ID: mdl-37099734

ABSTRACT

Hsp104 propagates the yeast prion [PSI+], the infectious form of Sup35, by severing the prion seeds, but when Hsp104 is overexpressed, it cures [PSI+] in a process that is not yet understood but may be caused by trimming, which removes monomers from the ends of the amyloid fibers. This curing was shown to depend on both the N-terminal domain of Hsp104 and the expression level of various members of the Hsp70 family, which raises the question as to whether these effects of Hsp70 are due to it binding to the Hsp70 binding site that was identified in the N-terminal domain of Hsp104, a site not involved in prion propagation. Investigating this question, we now find, first, that mutating this site prevents both the curing of [PSI+] by Hsp104 overexpression and the trimming activity of Hsp104. Second, we find that depending on the specific member of the Hsp70 family binding to the N-terminal domain of Hsp104, both trimming and the curing caused by Hsp104 overexpression are either increased or decreased in parallel. Therefore, the binding of Hsp70 to the N-terminal domain of Hsp104 regulates both the rate of [PSI+] trimming by Hsp104 and the rate of [PSI+] curing by Hsp104 overexpression.


Subject(s)
Prions , Saccharomyces cerevisiae Proteins , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Prions/genetics , Prions/metabolism , Peptide Termination Factors/chemistry
6.
Viruses ; 14(11)2022 10 25.
Article in English | MEDLINE | ID: mdl-36366434

ABSTRACT

Prions replicate by a self-templating mechanism. Infidelity in the process can lead to the emergence of new infectious structures, referred to as variants or strains. The question of whether prions are prone to mis-templating is not completely answered. Our previous experiments with 23 variants of the yeast [PSI+] prion do not support broad mutability. However, it became clear recently that the heat shock protein Hsp104 can restrict [PSI+] strain variation. This raises the possibility that many transmutable variants of the prion may have been mistaken as faithful-propagating simply because the mutant structure was too sturdy or too frail to take root in the wild-type cell. Here, I alter the strength of Hsp104 in yeast, overexpressing wild-type Hsp104 or expressing the hypo-active Hsp104T160M mutant, and check if the new environments enable the variants to mutate. Two variants hitherto thought of as faithful-propagating are discovered to generate different structures, which are stabilized with the hypo-active chaperone. In contrast, most transmutable variants discovered in cells overexpressing Hsp104 have been correctly identified as such previously in wild-type cells without the overexpression. The majority of transmutable variants only mis-template the structure of VH, VK, or VL, which are the most frequently observed variants and do not spontaneously mutate. There are four additional variants that never give rise to different structures in all cell conditions tested. Therefore, quite a few [PSI+] variants are faithful-propagating, and even the transmutable ones do not freely evolve but can only change to limited structural types.


Subject(s)
Prions , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Prions/genetics , Prions/metabolism , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism
7.
J Clin Invest ; 132(16)2022 08 15.
Article in English | MEDLINE | ID: mdl-35763353

ABSTRACT

Targeted protein degradation is a rapidly advancing and expanding therapeutic approach. Drugs that degrade GSPT1 via the CRL4CRBN ubiquitin ligase are a new class of cancer therapy in active clinical development with evidence of activity against acute myeloid leukemia in early-phase trials. However, other than activation of the integrated stress response, the downstream effects of GSPT1 degradation leading to cell death are largely undefined, and no murine models are available to study these agents. We identified the domains of GSPT1 essential for cell survival and show that GSPT1 degradation leads to impaired translation termination, activation of the integrated stress response pathway, and TP53-independent cell death. CRISPR/Cas9 screens implicated decreased translation initiation as protective following GSPT1 degradation, suggesting that cells with higher levels of translation are more susceptible to the effects of GSPT1 degradation. We defined 2 Crbn amino acids that prevent Gspt1 degradation in mice, generated a knockin mouse with alteration of these residues, and demonstrated the efficacy of GSPT1-degrading drugs in vivo with relative sparing of numbers and function of long-term hematopoietic stem cells. Our results provide a mechanistic basis for the use of GSPT1 degraders for the treatment of cancer, including TP53-mutant acute myeloid leukemia.


Subject(s)
Leukemia , Peptide Termination Factors , Animals , Cell Death , Hematopoietic Stem Cells/metabolism , Mice , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Proteolysis
8.
RNA Biol ; 18(sup2): 804-817, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34793288

ABSTRACT

Nsp1 of SARS-CoV-2 regulates the translation of host and viral mRNAs in cells. Nsp1 inhibits host translation initiation by occluding the entry channel of the 40S ribosome subunit. The structural study of the Nsp1-ribosomal complexes reported post-termination 80S complex containing Nsp1, eRF1 and ABCE1. Considering the presence of Nsp1 in the post-termination 80S ribosomal complex, we hypothesized that Nsp1 may be involved in translation termination. Using a cell-free translation system and reconstituted in vitro translation system, we show that Nsp1 stimulates peptide release and formation of termination complexes. Detailed analysis of Nsp1 activity during translation termination stages reveals that Nsp1 facilitates stop codon recognition. We demonstrate that Nsp1 stimulation targets eRF1 and does not affect eRF3. Moreover, Nsp1 increases amount of the termination complexes at all three stop codons. The activity of Nsp1 in translation termination is provided by its N-terminal domain and the minimal required part of eRF1 is NM domain. We assume that the biological meaning of Nsp1 activity in translation termination is binding with the 80S ribosomes translating host mRNAs and remove them from the pool of the active ribosomes.


Subject(s)
Protein Biosynthesis , SARS-CoV-2 , Viral Nonstructural Proteins/physiology , Animals , Cell-Free System , Codon, Terminator/metabolism , GTP Phosphohydrolases/metabolism , HeLa Cells , Humans , Mutation , Peptide Chain Termination, Translational , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Domains , RNA, Messenger/metabolism , Rabbits , Ribosomes/metabolism
9.
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
10.
J Biol Chem ; 296: 100681, 2021.
Article in English | MEDLINE | ID: mdl-33887323

ABSTRACT

Accurate translation termination in bacteria requires correct recognition of the stop codons by the class-I release factors (RFs) RF1 and RF2, which release the nascent peptide from the peptidyl tRNA after undergoing a "compact to open" conformational transition. These RFs possess a conserved Gly-Gly-Gln (GGQ) peptide release motif, of which the Q residue is posttranslationally methylated. GGQ-methylated RFs have been shown to be faster in peptide release than the unmethylated ones, but it was unknown whether this modification had additional roles. Using a fluorescence-based real-time in vitro translation termination assay in a stopped-flow instrument, we demonstrate that methylated RF1 and RF2 are two- to four-fold more accurate in the cognate stop codon recognition than their unmethylated variants. Using pH titration, we show that the lack of GGQ methylation facilitates the "compact to open" transition, which results in compromised accuracy of the unmethylated RFs. Furthermore, thermal melting studies using circular dichroism and SYPRO-orange fluorescence demonstrate that GGQ methylation increases overall stability of the RF proteins. This increased stability, we suspect, is the basis for the more controlled conformational change of the methylated RFs upon codon recognition, which enhances both their speed and accuracy. This GGQ methylation-based modulation of the accuracy of RFs can be a tool for regulating translational termination in vivo.


Subject(s)
Codon, Terminator/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Amino Acid Motifs , Conserved Sequence , Kinetics , Methylation , Models, Molecular , Temperature
11.
J Med Chem ; 64(4): 1835-1843, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33591756

ABSTRACT

Acute myeloid leukemia (AML) is marked by significant unmet clinical need due to both poor survival and high relapse rates where long-term disease control for most patients with relapsed or refractory AML remain dismal. Inspired to bring novel therapeutic options to these patients, we envisioned protein degradation as a potential therapeutic approach for the treatment of AML. Following this course, we discovered and pioneered a novel mechanism of action which culminated in the discovery of CC-90009. CC-90009 represents a novel protein degrader and the first cereblon E3 ligase modulating drug to enter clinical development that specifically targets GSPT1 (G1 to S phase transition 1) for proteasomal degradation. This manuscript briefly summarizes the mechanism of action, scientific rationale, medicinal chemistry, pharmacokinetic properties, and efficacy data for CC-90009, which is currently in phase 1 clinical development.


Subject(s)
Acetamides/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/therapeutic use , Isoindoles/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Peptide Termination Factors/antagonists & inhibitors , Piperidones/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Acetamides/chemistry , Acetamides/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Isoindoles/chemistry , Isoindoles/pharmacology , Macaca fascicularis , Male , Mice , Molecular Structure , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Piperidones/chemistry , Piperidones/pharmacology , Proteolysis/drug effects , Structure-Activity Relationship
12.
Biochemistry ; 59(48): 4533-4545, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33231432

ABSTRACT

RNA thermosensors (RNATs), found in the 5' untranslated region (UTR) of some bacterial messenger RNAs (mRNAs), control the translation of the downstream gene in a temperature-dependent manner. In Listeria monocytogenes, the expression of a key transcription factor, PrfA, is mediated by an RNAT in its 5' UTR. PrfA functions as a master regulator of virulence in L. monocytogenes, controlling the expression of many virulence factors. The temperature-regulated expression of PrfA by its RNAT element serves as a signal of successful host invasion for the bacteria. Structurally, the prfA RNAT bears little resemblance to known families of RNATs, and prior studies demonstrated that the prfA RNAT is highly responsive over a narrow temperature range. Herein, we have undertaken a comprehensive mutational and thermodynamic analysis to ascertain the molecular determinants of temperature sensitivity. We provide evidence to support the idea that the prfA RNAT unfolding is different from that of cssA, a well-characterized RNAT, suggesting that these RNATs function via distinct mechanisms. Our data show that the unfolding of the prfA RNAT occurs in two distinct events and that the internal loops play an important role in mediating the cooperativity of RNAT unfolding. We further demonstrated that regions distal to the ribosome binding site (RBS) not only contribute to RNAT structural stability but also impact translation of the downstream message. Our collective results provide insight connecting the thermal stability of the prfA RNAT structure, unfolding energetics, and translational control.


Subject(s)
Bacterial Proteins/chemistry , Peptide Termination Factors/chemistry , RNA, Bacterial/chemistry , 5' Untranslated Regions , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Gene Expression Regulation, Bacterial , Listeria monocytogenes/chemistry , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Magnesium/metabolism , Mutation , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Nucleic Acid Denaturation , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Potassium/metabolism , RNA Stability , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Temperature , Thermodynamics
13.
Proc Natl Acad Sci U S A ; 117(47): 29677-29683, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33168711

ABSTRACT

Yeast prions provide self-templating protein-based mechanisms of inheritance whose conformational changes lead to the acquisition of diverse new phenotypes. The best studied of these is the prion domain (NM) of Sup35, which forms an amyloid that can adopt several distinct conformations (strains) that confer distinct phenotypes when introduced into cells that do not carry the prion. Classic dyes, such as thioflavin T and Congo red, exhibit large increases in fluorescence when bound to amyloids, but these dyes are not sensitive to local structural differences that distinguish amyloid strains. Here we describe the use of Michler's hydrol blue (MHB) to investigate fibrils formed by the weak and strong prion fibrils of Sup35NM and find that MHB differentiates between these two polymorphs. Quantum mechanical time-dependent density functional theory (TDDFT) calculations indicate that the fluorescence properties of amyloid-bound MHB can be correlated to the change of binding site polarity and that a tyrosine to phenylalanine substitution at a binding site could be detected. Through the use of site-specific mutants, we demonstrate that MHB is a site-specific environmentally sensitive probe that can provide structural details about amyloid fibrils and their polymorphs.


Subject(s)
Aniline Compounds/chemistry , Benzhydryl Compounds/chemistry , Prions/chemistry , Amyloid/chemistry , Binding Sites , Congo Red/chemistry , Fluorescence , Fungal Proteins/chemistry , Peptide Termination Factors/chemistry , Protein Domains , Yeasts/chemistry
14.
Science ; 370(6520): 1105-1110, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33243891

ABSTRACT

The human mitochondrial ribosome (mitoribosome) and associated proteins regulate the synthesis of 13 essential subunits of the oxidative phosphorylation complexes. We report the discovery of a mitoribosome-associated quality control pathway that responds to interruptions during elongation, and we present structures at 3.1- to 3.3-angstrom resolution of mitoribosomal large subunits trapped during ribosome rescue. Release factor homolog C12orf65 (mtRF-R) and RNA binding protein C6orf203 (MTRES1) eject the nascent chain and peptidyl transfer RNA (tRNA), respectively, from stalled ribosomes. Recruitment of mitoribosome biogenesis factors to these quality control intermediates suggests additional roles for these factors during mitoribosome rescue. We also report related cryo-electron microscopy structures (3.7 to 4.4 angstrom resolution) of elongating mitoribosomes bound to tRNAs, nascent polypeptides, the guanosine triphosphatase elongation factors mtEF-Tu and mtEF-G1, and the Oxa1L translocase.


Subject(s)
Mitochondrial Ribosomes/chemistry , Transcription Elongation, Genetic , Cryoelectron Microscopy , Electron Transport Complex IV/chemistry , Escherichia coli , Exoribonucleases/genetics , HEK293 Cells , Humans , Mitochondrial Proteins/chemistry , Nuclear Proteins/chemistry , Peptide Termination Factors/chemistry , Protein Domains , RNA, Transfer/chemistry , RNA-Binding Proteins/chemistry , Ribosomal Proteins/chemistry , Transcriptional Elongation Factors/chemistry
15.
ACS Chem Biol ; 15(10): 2722-2730, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32865967

ABSTRACT

Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of neo-substrate proteins. In order to identify additional small molecule CRBN modulators, we implemented a focused combinatorial library approach where we fused an imide-based CRBN-binding pharmacophore to a heterocyclic scaffold, which could be further elaborated. We screened the library for CRBN-dependent antiproliferative activity in the multiple myeloma cell line MM1.S and identified five hit compounds. Quantitative chemical proteomics of hit compounds revealed that they induced selective degradation of GSPT1, a translation termination factor that is currently being explored as a therapeutic target for the treatment of acute myeloid leukemia. Molecular docking studies with CRBN and GSPT1 followed by analogue synthesis identified a possible hydrogen bond interaction with the central pyrimidine ring as a molecular determinant of hit compounds' selectivity. This study demonstrates that a focused combinatorial library design, phenotypic screening, and chemical proteomics can provide a suitable workflow to efficiently identify novel CRBN modulators.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Peptide Termination Factors/metabolism , Proteolysis/drug effects , Small Molecule Libraries/pharmacology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hydrogen Bonding , Molecular Docking Simulation , Peptide Termination Factors/chemistry , Protein Binding , Small Molecule Libraries/metabolism , Thalidomide/metabolism , Ubiquitin-Protein Ligases/chemistry
16.
Biochem Biophys Res Commun ; 529(3): 533-539, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32736670

ABSTRACT

The translation terminator Sup35p assembles into self-replicating fibrillar aggregates that are responsible for the [PSI+] prion state. The Q/N-rich N-terminal domain together with the highly charged middle-domain (NM domain) drive the assembly of Sup35p into amyloid fibrils in vitro. NM domains are highly divergent among yeasts. The ability to convert to a prion form is however conserved among Sup35 orthologs. In particular, the Yarrowia lipolytica Sup35p stands out with an exceptionally high prion conversion rate. In the present work, we show that different Yarrowia lipolytica strains contain one of two Sup35p orthologs that differ by the number of repeats within their NM domain. The Y. lipolytica Sup35 proteins are able to assemble into amyloid fibrils. Contrary to S. cerevisiae Sup35p, fibrils made of full-length or NM domains of Y. lipolytica Sup35 proteins did not bind Thioflavin-T, a well-known marker of amyloid aggregates.


Subject(s)
Amyloid/metabolism , Benzothiazoles/metabolism , Fungal Proteins/metabolism , Prions/metabolism , Yarrowia/metabolism , Amino Acid Sequence , Amyloid/chemistry , Amyloid/ultrastructure , Fungal Proteins/chemistry , Fungal Proteins/genetics , Microscopy, Electron, Transmission , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Prions/chemistry , Prions/genetics , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Yarrowia/genetics
17.
Nat Struct Mol Biol ; 27(6): 540-549, 2020 06.
Article in English | MEDLINE | ID: mdl-32367069

ABSTRACT

Amyloid appearance is a rare event that is promoted in the presence of other aggregated proteins. These aggregates were thought to act by templating the formation of an assembly-competent nucleation seed, but we find an unanticipated role for them in enhancing the persistence of amyloid after it arises. Specifically, Saccharomyces cerevisiae Rnq1 amyloid reduces chaperone-mediated disassembly of Sup35 amyloid, promoting its persistence in yeast. Mathematical modeling and corresponding in vivo experiments link amyloid persistence to the conformationally defined size of the Sup35 nucleation seed and suggest that amyloid is actively cleared by disassembly below this threshold to suppress appearance of the [PSI+] prion in vivo. Remarkably, this framework resolves multiple known inconsistencies in the appearance and curing of yeast prions. Thus, our observations establish the size of the nucleation seed as a previously unappreciated characteristic of prion variants that is key to understanding transitions between prion states.


Subject(s)
Amyloid/metabolism , Heat-Shock Proteins/metabolism , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Amyloid/chemistry , Cycloheximide/pharmacology , Heat-Shock Proteins/chemistry , Peptide Termination Factors/genetics , Prions/chemistry , Prions/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
18.
Nat Chem Biol ; 16(7): 756-765, 2020 07.
Article in English | MEDLINE | ID: mdl-32284601

ABSTRACT

Soluble prion proteins contingently encounter foreign prion aggregates, leading to cross-species prion transmission. However, how its efficiency is regulated by structural fluctuation of the host soluble prion protein remains unsolved. In the present study, through the use of two distantly related yeast prion Sup35 proteins, we found that a specific conformation of a short disordered segment governs interspecies prion transmissibility. Using a multidisciplinary approach including high-resolution NMR and molecular dynamics simulation, we identified critical residues within this segment that allow interspecies prion transmission in vitro and in vivo, by locally altering dynamics and conformation of soluble prion proteins. Remarkably, subtle conformational differences caused by a methylene group between asparagine and glutamine sufficed to change the short segment structure and substantially modulate the cross-seeding activity. Thus, our findings uncover how conformational dynamics of the short segment in the host prion protein impacts cross-species prion transmission. More broadly, our study provides mechanistic insights into cross-seeding between heterologous proteins.


Subject(s)
Asparagine/chemistry , Glutamine/chemistry , Intrinsically Disordered Proteins/chemistry , Peptide Termination Factors/chemistry , Prions/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Asparagine/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutamine/metabolism , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Molecular Dynamics Simulation , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Prions/genetics , Prions/metabolism , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics
19.
Proc Natl Acad Sci U S A ; 117(14): 7831-7836, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32213585

ABSTRACT

The yeast prion protein Sup35, which contains intrinsically disordered regions, forms amyloid fibrils responsible for a prion phenotype [PSI+]. Using high-speed atomic force microscopy (HS-AFM), we directly visualized the prion determinant domain (Sup35NM) and the formation of its oligomers and fibrils at subsecond and submolecular resolutions. Monomers with freely moving tail-like regions initially appeared in the images, and subsequently oligomers with distinct sizes of ∼1.7 and 3 to 4 nm progressively accumulated. Nevertheless, these oligomers did not form fibrils, even after an incubation for 2 h in the presence of monomers. Fibrils appeared after much longer monomer incubation. The fibril elongation occurred smoothly without discrete steps, suggesting gradual conversions of the incorporated monomers into cross-ß structures. The individual oligomers were separated from each other and also from the fibrils by respective, identical lengths on the mica surface, probably due to repulsion caused by the freely moving disordered regions. Based on these HS-AFM observations, we propose that the freely moving tails of the monomers are incorporated into the fibril ends, and then the structural conversions to cross-ß structures gradually occur.


Subject(s)
Amyloid/ultrastructure , Microscopy, Atomic Force , Peptide Termination Factors/ultrastructure , Prion Proteins/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae/ultrastructure , Amyloid/genetics , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Prion Proteins/genetics , Protein Conformation, beta-Strand/genetics , Protein Domains/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
20.
Nat Microbiol ; 5(4): 554-561, 2020 04.
Article in English | MEDLINE | ID: mdl-32094585

ABSTRACT

Polyamines are essential metabolites that play an important role in cell growth, stress adaptation and microbial virulence1-3. To survive and multiply within a human host, pathogenic bacteria adjust the expression and activity of polyamine biosynthetic enzymes in response to different environmental stresses and metabolic cues2. Here, we show that ornithine capture by the ribosome and the nascent peptide SpeFL controls polyamine synthesis in γ-proteobacteria by inducing the expression of the ornithine decarboxylase SpeF4, via a mechanism involving ribosome stalling and transcription antitermination. In addition, we present the cryogenic electron microscopy structure of an Escherichia coli ribosome stalled during translation of speFL in the presence of ornithine. The structure shows how the ribosome and the SpeFL sensor domain form a highly selective binding pocket that accommodates a single ornithine molecule but excludes near-cognate ligands. Ornithine pre-associates with the ribosome and is then held in place by the sensor domain, leading to the compaction of the SpeFL effector domain and blocking the action of release factor 1. Thus, our study not only reveals basic strategies by which nascent peptides assist the ribosome in detecting a specific metabolite, but also provides a framework for assessing how ornithine promotes virulence in several human pathogens.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli/genetics , Ornithine Decarboxylase/chemistry , Ornithine/chemistry , Ribosomes/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Binding Sites , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Models, Molecular , Ornithine/metabolism , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Phylogeny , Polyamines/chemistry , Polyamines/metabolism , Protein Binding , Protein Biosynthesis , Protein Interaction Domains and Motifs , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ribosomes/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Sequence Alignment , Sequence Homology, Amino Acid , Thermus thermophilus/genetics , Thermus thermophilus/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...