Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
1.
Biochemistry (Mosc) ; 87(9): 871-889, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36180983

ABSTRACT

Design and synthesis of conjugates consisting of the macrolide antibiotic desmycosin and fragments of the antibacterial peptide oncocin were performed in attempt to develop new antimicrobial compounds. New compounds were shown to bind to the E. coli 70S ribosomes, to inhibit bacterial protein synthesis in vitro, as well as to suppress bacterial growth. The conjugates of N-terminal hexa- and tripeptide fragments of oncocin and 3,2',4''-triacetyldesmycosin were found to be active against some strains of macrolide-resistant bacteria. By simulating molecular dynamics of the complexes of these compounds with the wild-type bacterial ribosomes and with ribosomes, containing A2059G 23S RNA mutation, the specific structural features of their interactions were revealed.


Subject(s)
Antimicrobial Peptides , Escherichia coli , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Macrolides/analysis , Macrolides/metabolism , Protein Synthesis Inhibitors/chemistry , RNA/metabolism , Ribosomes/chemistry , Tylosin/analogs & derivatives
3.
Nat Struct Mol Biol ; 29(2): 152-161, 2022 02.
Article in English | MEDLINE | ID: mdl-35165455

ABSTRACT

Ribosome-targeting antibiotics serve as powerful antimicrobials and as tools for studying the ribosome, the catalytic peptidyl transferase center (PTC) of which is targeted by many drugs. The classic PTC-acting antibiotic chloramphenicol (CHL) and the newest clinically significant linezolid (LZD) were considered indiscriminate inhibitors of protein synthesis that cause ribosome stalling at every codon of every gene being translated. However, recent discoveries have shown that CHL and LZD preferentially arrest translation when the ribosome needs to polymerize particular amino acid sequences. The molecular mechanisms that underlie the context-specific action of ribosome inhibitors are unknown. Here we present high-resolution structures of ribosomal complexes, with or without CHL, carrying specific nascent peptides that support or negate the drug action. Our data suggest that the penultimate residue of the nascent peptide directly modulates antibiotic affinity to the ribosome by either establishing specific interactions with the drug or by obstructing its proper placement in the binding site.


Subject(s)
Chloramphenicol/chemistry , Chloramphenicol/pharmacology , Peptidyl Transferases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Models, Molecular , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Static Electricity , Thermus thermophilus/drug effects , Thermus thermophilus/metabolism
4.
Eur J Med Chem ; 226: 113856, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34547506

ABSTRACT

Retinol-binding protein 4 (RBP4) is a serum protein that transports Vitamin A. RBP4 is correlated with numerous diseases and metabolic syndromes, including insulin resistance in type 2 diabetes, cardiovascular diseases, obesity, and macular degeneration. Recently, RBP4 antagonists and protein synthesis inhibitors are under development to regulate the effect of RBP4. Several RBP4 antagonists, especially BPN-14136, have demonstrated promising safety profiles and potential therapeutic benefits in animal studies. Two RBP4 antagonists, specifically tinlarebant (Belite Bio) and STG-001 (Stargazer) are currently undergoing clinical trials. Some antidiabetic drugs and nutraceuticals have been reported to reduce RBP4 expression, but more clinical data is needed to evaluate their therapeutical benefits. As regulating RBP4 levels or its activities would benefit a wide range of patients, further research is highly recommended to develop clinically useful RBP4 antagonists or protein synthesis inhibitors.


Subject(s)
Carboxylic Acids/pharmacology , Drug Development , Protein Synthesis Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Retinol-Binding Proteins, Plasma/antagonists & inhibitors , Carboxylic Acids/chemical synthesis , Carboxylic Acids/chemistry , Humans , Protein Synthesis Inhibitors/chemical synthesis , Protein Synthesis Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Retinol-Binding Proteins, Plasma/biosynthesis
5.
Org Lett ; 23(17): 6967-6971, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34388000

ABSTRACT

A cryptic trans-acyltransferase polyketide synthase biosynthetic gene cluster sdl (80 kb) from Streptomyces sp. B59 was cloned and transferred into a heterologous host Streptomyces albus J1074, resulting in a class of polycyclic macrolide shuangdaolides A-D (1-4) and dumulmycin (5). Heterologous expression and gene inactivation experiments allowed the identification of two biosynthetic intermediates, 6 and 7, suggesting an unusual multidomain SDR oxidoreductase SdlR in charge of the formation of a rare 2-hydroxycyclopentenone moiety in this class of compounds.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Macrolides/chemistry , Polyketide Synthases/chemistry , Protein Synthesis Inhibitors/chemistry , Streptomyces/chemistry , Anti-Bacterial Agents/chemistry , Macrolides/metabolism , Molecular Structure , Multigene Family , Polyketide Synthases/metabolism , Protein Synthesis Inhibitors/metabolism
6.
Org Lett ; 23(18): 7106-7111, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34436915

ABSTRACT

Saccharothriolides A-F are 10-membered microbial macrolides proposed to be generated from their precursors presaccharothriolides X-Z. Previously, we isolated presaccharothriolide X, and its unique natural prodrug-like properties have intrigued us. However, the other congeners were not detected. Herein, we detected presaccharothriolide Z using our highly sensitive labeling reagent. Moreover, chemical synthesis of presaccharothriolide Z, the first total synthesis of saccharothriolide-class macrolides, was achieved, and the structure and biological activity of presaccharothriolide Z were determined.


Subject(s)
Actinomycetales/chemistry , Anti-Bacterial Agents/chemical synthesis , Macrolides/chemical synthesis , Protein Synthesis Inhibitors/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Macrolides/chemistry , Macrolides/pharmacology , Molecular Structure
7.
Nat Commun ; 12(1): 4466, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294725

ABSTRACT

Macrolides and ketolides comprise a family of clinically important antibiotics that inhibit protein synthesis by binding within the exit tunnel of the bacterial ribosome. While these antibiotics are known to interrupt translation at specific sequence motifs, with ketolides predominantly stalling at Arg/Lys-X-Arg/Lys motifs and macrolides displaying a broader specificity, a structural basis for their context-specific action has been lacking. Here, we present structures of ribosomes arrested during the synthesis of an Arg-Leu-Arg sequence by the macrolide erythromycin (ERY) and the ketolide telithromycin (TEL). Together with deep mutagenesis and molecular dynamics simulations, the structures reveal how ERY and TEL interplay with the Arg-Leu-Arg motif to induce translational arrest and illuminate the basis for the less stringent sequence-specific action of ERY over TEL. Because programmed stalling at the Arg/Lys-X-Arg/Lys motifs is used to activate expression of antibiotic resistance genes, our study also provides important insights for future development of improved macrolide antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ketolides/pharmacology , Macrolides/pharmacology , Protein Synthesis Inhibitors/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Bacillus subtilis/drug effects , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Binding Sites/genetics , Cryoelectron Microscopy , Drug Resistance, Microbial/genetics , Erythromycin/chemistry , Erythromycin/pharmacology , Genes, Bacterial , Ketolides/chemistry , Ketolides/pharmacokinetics , Macrolides/chemistry , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Molecular Dynamics Simulation , Mutagenesis, Insertional , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , Ribosomes/drug effects
8.
Nucleic Acids Res ; 49(13): 7665-7679, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34157102

ABSTRACT

Deciphering translation is of paramount importance for the understanding of many diseases, and antibiotics played a pivotal role in this endeavour. Blasticidin S (BlaS) targets translation by binding to the peptidyl transferase center of the large ribosomal subunit. Using biochemical, structural and cellular approaches, we show here that BlaS inhibits both translation elongation and termination in Mammalia. Bound to mammalian terminating ribosomes, BlaS distorts the 3'CCA tail of the P-site tRNA to a larger extent than previously reported for bacterial ribosomes, thus delaying both, peptide bond formation and peptidyl-tRNA hydrolysis. While BlaS does not inhibit stop codon recognition by the eukaryotic release factor 1 (eRF1), it interferes with eRF1's accommodation into the peptidyl transferase center and subsequent peptide release. In human cells, BlaS inhibits nonsense-mediated mRNA decay and, at subinhibitory concentrations, modulates translation dynamics at premature termination codons leading to enhanced protein production.


Subject(s)
Peptide Chain Elongation, Translational/drug effects , Peptide Chain Termination, Translational/drug effects , Protein Synthesis Inhibitors/pharmacology , Cryoelectron Microscopy , HeLa Cells , Humans , Nonsense Mediated mRNA Decay/drug effects , Nucleosides/chemistry , Nucleosides/pharmacology , Peptide Termination Factors/metabolism , Peptides/metabolism , Protein Synthesis Inhibitors/chemistry , RNA, Messenger/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Large, Eukaryotic/drug effects , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/metabolism
9.
Nucleic Acids Res ; 49(12): 6880-6892, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34125898

ABSTRACT

How aminoglycoside antibiotics limit bacterial growth and viability is not clearly understood. Here we employ fast kinetics to reveal the molecular mechanism of action of a clinically used, new-generation, semisynthetic aminoglycoside Arbekacin (ABK), which is designed to avoid enzyme-mediated deactivation common to other aminoglycosides. Our results portray complete picture of ABK inhibition of bacterial translation with precise quantitative characterizations. We find that ABK inhibits different steps of translation in nanomolar to micromolar concentrations by imparting pleotropic effects. ABK binding stalls elongating ribosomes to a state, which is unfavorable for EF-G binding. This prolongs individual translocation step from ∼50 ms to at least 2 s; the mean time of translocation increases inversely with EF-G concentration. ABK also inhibits translation termination by obstructing RF1/RF2 binding to the ribosome. Furthermore, ABK decreases accuracy of mRNA decoding (UUC vs. CUC) by ∼80 000 fold, causing aberrant protein production. Importantly, translocation and termination events cannot be completely stopped even with high ABK concentration. Extrapolating our kinetic model of ABK action, we postulate that aminoglycosides impose bacteriostatic effect mainly by inhibiting translocation, while they become bactericidal in combination with decoding errors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dibekacin/analogs & derivatives , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Ribosomes/drug effects , Anti-Bacterial Agents/chemistry , Dibekacin/chemistry , Dibekacin/pharmacology , Kinetics , Peptide Elongation Factor G/antagonists & inhibitors , Peptide Termination Factors/antagonists & inhibitors , Peptides/metabolism , Protein Synthesis Inhibitors/chemistry , RNA, Messenger/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/metabolism
10.
Nat Commun ; 12(1): 2803, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990576

ABSTRACT

Macrolide antibiotics bind in the nascent peptide exit tunnel of the bacterial ribosome and prevent polymerization of specific amino acid sequences, selectively inhibiting translation of a subset of proteins. Because preventing translation of individual proteins could be beneficial for the treatment of human diseases, we asked whether macrolides, if bound to the eukaryotic ribosome, would retain their context- and protein-specific action. By introducing a single mutation in rRNA, we rendered yeast Saccharomyces cerevisiae cells sensitive to macrolides. Cryo-EM structural analysis showed that the macrolide telithromycin binds in the tunnel of the engineered eukaryotic ribosome. Genome-wide analysis of cellular translation and biochemical studies demonstrated that the drug inhibits eukaryotic translation by preferentially stalling ribosomes at distinct sequence motifs. Context-specific action markedly depends on the macrolide structure. Eliminating macrolide-arrest motifs from a protein renders its translation macrolide-tolerant. Our data illuminate the prospects of adapting macrolides for protein-selective translation inhibition in eukaryotic cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Macrolides/pharmacology , Ribosomes/drug effects , Anti-Bacterial Agents/chemistry , Binding Sites , Cryoelectron Microscopy , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Humans , Macrolides/chemistry , Models, Molecular , Mutation , Protein Binding , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Fungal/genetics , RNA, Ribosomal/genetics , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Structure-Activity Relationship
11.
Nat Commun ; 12(1): 1799, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741965

ABSTRACT

Bacterial ribosome rescue pathways that remove ribosomes stalled on mRNAs during translation have been proposed as novel antibiotic targets because they are essential in bacteria and are not conserved in humans. We previously reported the discovery of a family of acylaminooxadiazoles that selectively inhibit trans-translation, the main ribosome rescue pathway in bacteria. Here, we report optimization of the pharmacokinetic and antibiotic properties of the acylaminooxadiazoles, producing MBX-4132, which clears multiple-drug resistant Neisseria gonorrhoeae infection in mice after a single oral dose. Single particle cryogenic-EM studies of non-stop ribosomes show that acylaminooxadiazoles bind to a unique site near the peptidyl-transfer center and significantly alter the conformation of ribosomal protein bL27, suggesting a novel mechanism for specific inhibition of trans-translation by these molecules. These results show that trans-translation is a viable therapeutic target and reveal a new conformation within the bacterial ribosome that may be critical for ribosome rescue pathways.


Subject(s)
Neisseria gonorrhoeae/drug effects , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Ribosomes/drug effects , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites/genetics , Caco-2 Cells , Female , Gonorrhea/microbiology , Gonorrhea/prevention & control , Humans , Mice , Neisseria gonorrhoeae/genetics , Protein Biosynthesis/genetics , Protein Synthesis Inhibitors/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism
12.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33674389

ABSTRACT

Apidaecin (Api), an unmodified 18-amino-acid-long proline-rich antibacterial peptide produced by bees, has been recently described as a specific inhibitor of translation termination. It invades the nascent peptide exit tunnel of the postrelease ribosome and traps the release factors preventing their recycling. Api binds in the exit tunnel in an extended conformation that matches the placement of a nascent polypeptide and establishes multiple contacts with ribosomal RNA (rRNA) and ribosomal proteins. Which of these interactions are critical for Api's activity is unknown. We addressed this problem by analyzing the activity of all possible single-amino-acid substitutions of the Api variants synthesized in the bacterial cell. By conditionally expressing the engineered api gene, we generated Api directly in the bacterial cytosol, thereby bypassing the need for importing the peptide from the medium. The endogenously expressed Api, as well as its N-terminally truncated mutants, retained the antibacterial properties and the mechanism of action of the native peptide. Taking advantage of the Api expression system and next-generation sequencing, we mapped in one experiment all the single-amino-acid substitutions that preserve or alleviate the on-target activity of the Api mutants. Analysis of the inactivating mutations made it possible to define the pharmacophore of Api involved in critical interactions with the ribosome, transfer RNA (tRNA), and release factors. We also identified the Api segment that tolerates a variety of amino acid substitutions; alterations in this segment could be used to improve the pharmacological properties of the antibacterial peptide.


Subject(s)
Antimicrobial Cationic Peptides , Escherichia coli , Peptide Chain Termination, Translational/drug effects , Protein Synthesis Inhibitors , Amino Acid Substitution , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/pharmacology , Bees , Escherichia coli/genetics , Escherichia coli/metabolism , Mutation, Missense , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism
13.
Biochemistry (Mosc) ; 85(11): 1389-1421, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33280581

ABSTRACT

Eukaryotic ribosome and cap-dependent translation are attractive targets in the antitumor, antiviral, anti-inflammatory, and antiparasitic therapies. Currently, a broad array of small-molecule drugs is known that specifically inhibit protein synthesis in eukaryotic cells. Many of them are well-studied ribosome-targeting antibiotics that block translocation, the peptidyl transferase center or the polypeptide exit tunnel, modulate the binding of translation machinery components to the ribosome, and induce miscoding, premature termination or stop codon readthrough. Such inhibitors are widely used as anticancer, anthelmintic and antifungal agents in medicine, as well as fungicides in agriculture. Chemicals that affect the accuracy of stop codon recognition are promising drugs for the nonsense suppression therapy of hereditary diseases and restoration of tumor suppressor function in cancer cells. Other compounds inhibit aminoacyl-tRNA synthetases, translation factors, and components of translation-associated signaling pathways, including mTOR kinase. Some of them have antidepressant, immunosuppressive and geroprotective properties. Translation inhibitors are also used in research for gene expression analysis by ribosome profiling, as well as in cell culture techniques. In this article, we review well-studied and less known inhibitors of eukaryotic protein synthesis (with the exception of mitochondrial and plastid translation) classified by their targets and briefly describe the action mechanisms of these compounds. We also present a continuously updated database (http://eupsic.belozersky.msu.ru) that currently contains information on 370 inhibitors of eukaryotic protein synthesis.


Subject(s)
Anti-Bacterial Agents , Eukaryotic Cells/metabolism , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Humans , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/therapeutic use
14.
Org Lett ; 22(17): 6709-6713, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32808790

ABSTRACT

A novel family of four potent antimalarial macrolides, strasseriolides A-D (1-4), has been isolated from cultures of Strasseria geniculata CF-247251, a fungal strain obtained from plant tissues. The structures of these compounds, including their absolute configurations, were elucidated by HRMS, NMR spectroscopy, and X-ray single-crystal diffraction. The four compounds gave respective IC50 values of 9.810, 0.013, 0.123, and 0.128 µM against Plasmodium falciparum 3D7 parasites with no significant cytotoxicity against the HepG2 cell line.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimalarials/pharmacology , Macrolides/pharmacology , Protein Synthesis Inhibitors/analysis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antimalarials/chemistry , Antimalarials/isolation & purification , Ascomycota , Fungi , Macrolides/chemistry , Macrolides/isolation & purification , Molecular Structure , Protein Synthesis Inhibitors/chemistry
16.
J Biol Chem ; 295(29): 9855-9867, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32430400

ABSTRACT

Fluorizoline (FLZ) binds to prohibitin-1 and -2 (PHB1/2), which are pleiotropic scaffold proteins known to affect signaling pathways involved in several intracellular processes. However, it is not yet clear how FLZ exerts its effect. Here, we show that exposure of three different human cancer cell lines to FLZ increases the phosphorylation of key translation factors, particularly of initiation factor 2 (eIF2) and elongation factor 2 (eEF2), modifications that inhibit their activities. FLZ also impaired signaling through mTOR complex 1, which also regulates the translational machinery, e.g. through the eIF4E-binding protein 4E-BP1. In line with these findings, FLZ potently inhibited protein synthesis. We noted that the first phase of this inhibition involves very rapid eEF2 phosphorylation, which is catalyzed by a dedicated Ca2+-dependent protein kinase, eEF2 kinase (eEF2K). We also demonstrate that FLZ induces a swift and marked rise in intracellular Ca2+ levels, likely explaining the effects on eEF2. Disruption of normal Ca2+ homeostasis can also induce endoplasmic reticulum stress, and our results suggest that induction of this stress response contributes to the increased phosphorylation of eIF2, likely because of activation of the eIF2-modifying kinase PKR-like endoplasmic reticulum kinase (PERK). We show that FLZ induces cancer cell death and that this effect involves contributions from the phosphorylation of both eEF2 and eIF2. Our findings provide important new insights into the biological effects of FLZ and thus the roles of PHBs, specifically in regulating Ca2+ levels, cellular protein synthesis, and cell survival.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum Stress/drug effects , Neoplasm Proteins/biosynthesis , Neoplasms/metabolism , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , A549 Cells , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , HeLa Cells , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Peptide Elongation Factor 2/metabolism , Phosphorylation/drug effects , Prohibitins , Protein Synthesis Inhibitors/chemistry , Repressor Proteins/metabolism
17.
RNA ; 26(6): 715-723, 2020 06.
Article in English | MEDLINE | ID: mdl-32144191

ABSTRACT

Macrolides are one of the most successful and widely used classes of antibacterials, which kill or stop the growth of pathogenic bacteria by binding near the active site of the ribosome and interfering with protein synthesis. Dirithromycin is a derivative of the prototype macrolide erythromycin with additional hydrophobic side chain. In our recent study, we have discovered that the side chain of dirithromycin forms lone pair-π stacking interaction with the aromatic imidazole ring of the His69 residue in ribosomal protein uL4 of the Thermus thermophilus 70S ribosome. In the current work, we found that neither the presence of the side chain, nor the additional contact with the ribosome, improve the binding affinity of dirithromycin to the ribosome. Nevertheless, we found that dirithromycin is a more potent inhibitor of in vitro protein synthesis in comparison with its parent compound, erythromycin. Using high-resolution cryo-electron microscopy, we determined the structure of the dirithromycin bound to the translating Escherichia coli 70S ribosome, which suggests that the better inhibitory properties of the drug could be rationalized by the side chain of dirithromycin pointing into the lumen of the nascent peptide exit tunnel, where it can interfere with the normal passage of the growing polypeptide chain.


Subject(s)
Anti-Bacterial Agents/chemistry , Erythromycin/analogs & derivatives , Protein Synthesis Inhibitors/chemistry , Ribosomes/chemistry , Anti-Bacterial Agents/pharmacology , Cryoelectron Microscopy , Erythromycin/chemistry , Erythromycin/pharmacology , Escherichia coli/genetics , Models, Molecular , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Ribosomal, 23S/chemistry
18.
Structure ; 28(5): 528-539.e9, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32220302

ABSTRACT

Phenomycin is a bacterial mini-protein of 89 amino acids discovered more than 50 years ago with toxicity in the nanomolar regime toward mammalian cells. The protein inhibits the function of the eukaryotic ribosome in cell-free systems and appears to target translation initiation. Several fundamental questions concerning the cellular activity of phenomycin, however, have remained unanswered. In this paper, we have used morphological profiling to show that direct inhibition of translation underlies the toxicity of phenomycin in cells. We have performed studies of the cellular uptake mechanism of phenomycin, showing that endosomal escape is the toxicity-limiting step, and we have solved a solution phase high-resolution structure of the protein using NMR spectroscopy. Through bioinformatic as well as functional comparisons between phenomycin and two homologs, we have identified a peptide segment, which constitutes one of two loops in the structure that is critical for the toxicity of phenomycin.


Subject(s)
Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/toxicity , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Bacteriocins/pharmacokinetics , Bacteriocins/toxicity , Cell Line , Endosomes/drug effects , Endosomes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells , Mice , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/toxicity , Structure-Activity Relationship
19.
Proc Natl Acad Sci U S A ; 117(3): 1457-1467, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31900363

ABSTRACT

Many proteins are refractory to targeting because they lack small-molecule binding pockets. An alternative to drugging these proteins directly is to target the messenger (m)RNA that encodes them, thereby reducing protein levels. We describe such an approach for the difficult-to-target protein α-synuclein encoded by the SNCA gene. Multiplication of the SNCA gene locus causes dominantly inherited Parkinson's disease (PD), and α-synuclein protein aggregates in Lewy bodies and Lewy neurites in sporadic PD. Thus, reducing the expression of α-synuclein protein is expected to have therapeutic value. Fortuitously, the SNCA mRNA has a structured iron-responsive element (IRE) in its 5' untranslated region (5' UTR) that controls its translation. Using sequence-based design, we discovered small molecules that target the IRE structure and inhibit SNCA translation in cells, the most potent of which is named Synucleozid. Both in vitro and cellular profiling studies showed Synucleozid directly targets the α-synuclein mRNA 5' UTR at the designed site. Mechanistic studies revealed that Synucleozid reduces α-synuclein protein levels by decreasing the amount of SNCA mRNA loaded into polysomes, mechanistically providing a cytoprotective effect in cells. Proteome- and transcriptome-wide studies showed that the compound's selectivity makes Synucleozid suitable for further development. Importantly, transcriptome-wide analysis of mRNAs that encode intrinsically disordered proteins revealed that each has structured regions that could be targeted with small molecules. These findings demonstrate the potential for targeting undruggable proteins at the level of their coding mRNAs. This approach, as applied to SNCA, is a promising disease-modifying therapeutic strategy for PD and other α-synucleinopathies.


Subject(s)
Intrinsically Disordered Proteins/genetics , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Response Elements , alpha-Synuclein/genetics , 3' Untranslated Regions , Animals , Cell Line, Tumor , Humans , Intrinsically Disordered Proteins/metabolism , Mice , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , alpha-Synuclein/metabolism
20.
Phytomedicine ; 73: 152753, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-30773353

ABSTRACT

BACKGROUND: Fungal pathogenesis continues to be a burden to healthcare structures in both developed and developing nations. The gradual and irreversible loss of efficacies of existing antifungal medicines as well as the emergence of drug-resistant strains have contributed largely to this scenario. There is therefore a pressing need for new drugs from diverse structural backgrounds with improved potencies and novel modes of action to fortify or replace contemporary antifungal schedules. AIM: Alkaloids of the plant family Amaryllidaceae exhibit good growth inhibitory activities against several fungal pathogens. This review focuses on the mechanistic aspects of these antifungal activities. It achieves this by highlighting the molecular targets as well as structural features of Amaryllidaceae constituents which serve to enhance such action. METHODS: During the information gathering stage extensive use was made of the three database platforms; Google Scholar, SciFinder and Scopus. In most instances articles were accessed directly from journals licensed to the University of KwaZulu-Natal. In the absence of such proprietary agreements the respective corresponding authors were approached directly for copies of papers. RESULTS: Although several classes of molecules from the Amaryllidaceae have been probed for their antifungal effects, it is the key constituents lycorine and narciclasine which have together afforded the most profound mechanistic insights. These may be summarized as follows: (i) effects on the fungal cell wall and cell membrane; (ii) effects on morphology such as budding and hyphal growth; (iii) effects on fungal organelles such as ribosomes; (iv) effects on macromolecules such as DNA, RNA and proteins and; (v) identification of the active sites for these constituents. CONCLUSION: The key feature in the antifungal effects of Amaryllidaceae alkaloids is the inhibition of protein synthesis. This involved the inhibition of peptide bond formation by binding to yeast ribosomes via the 60S subunit. Related effects involved the inhibition of both DNA and RNA synthesis. These adverse effects were reflected morphologically on both the fungal cell wall and cell membrane. Such observations should prove useful in the chemotherapeutic arena should efforts shift towards the development of a clinical candidate.


Subject(s)
Amaryllidaceae/chemistry , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Alkaloids/chemistry , Alkaloids/pharmacology , Amaryllidaceae Alkaloids/pharmacology , Cell Wall/drug effects , Phenanthridines/pharmacology , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...