Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Curr Opin Microbiol ; 77: 102418, 2024 02.
Article in English | MEDLINE | ID: mdl-38159358

ABSTRACT

Sensing small molecules is crucial for microorganisms to adapt their genetic programs to changes in their environment. Arrest peptides encoded by short regulatory open reading frames program the ribosomes that translate them to undergo translational arrest in response to specific metabolites. Ribosome stalling in turn controls the expression of downstream genes on the same messenger RNA by translational or transcriptional means. In this review, we present our current understanding of the mechanisms by which ribosomes translating arrest peptides sense different metabolites, such as antibiotics or amino acids, to control gene expression.


Subject(s)
Protein Biosynthesis , Ribosomes , Ribosomes/genetics , Ribosomes/metabolism , Peptides/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism
2.
Nat Commun ; 14(1): 3891, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37393329

ABSTRACT

Antibiotic resistance ABC-Fs (ARE ABC-Fs) are translation factors that provide resistance against clinically important ribosome-targeting antibiotics which are proliferating among pathogens. Here, we combine genetic and structural approaches to determine the regulation of streptococcal ARE ABC-F gene msrD in response to macrolide exposure. We show that binding of cladinose-containing macrolides to the ribosome prompts insertion of the leader peptide MsrDL into a crevice of the ribosomal exit tunnel, which is conserved throughout bacteria and eukaryotes. This leads to a local rearrangement of the 23 S rRNA that prevents peptide bond formation and accommodation of release factors. The stalled ribosome obstructs the formation of a Rho-independent terminator structure that prevents msrD transcriptional attenuation. Erythromycin induction of msrD expression via MsrDL, is suppressed by ectopic expression of mrsD, but not by mutants which do not provide antibiotic resistance, showing correlation between MsrD function in antibiotic resistance and its action on this stalled complex.


Subject(s)
Abducens Nerve Diseases , Anti-Bacterial Agents , Humans , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Macrolides/pharmacology , Accommodation, Ocular
3.
Nat Chem Biol ; 19(9): 1091-1096, 2023 09.
Article in English | MEDLINE | ID: mdl-37322159

ABSTRACT

As antimicrobial resistance threatens our ability to treat common bacterial infections, new antibiotics with limited cross-resistance are urgently needed. In this regard, natural products that target the bacterial ribosome have the potential to be developed into potent drugs through structure-guided design, provided their mechanisms of action are well understood. Here we use inverse toeprinting coupled to next-generation sequencing to show that the aromatic polyketide tetracenomycin X primarily inhibits peptide bond formation between an incoming aminoacyl-tRNA and a terminal Gln-Lys (QK) motif in the nascent polypeptide. Using cryogenic electron microscopy, we reveal that translation inhibition at QK motifs occurs via an unusual mechanism involving sequestration of the 3' adenosine of peptidyl-tRNALys in the drug-occupied nascent polypeptide exit tunnel of the ribosome. Our study provides mechanistic insights into the mode of action of tetracenomycin X on the bacterial ribosome and suggests a path forward for the development of novel aromatic polyketide antibiotics.


Subject(s)
Anti-Bacterial Agents , Polyketides , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Peptides/pharmacology , Peptides/chemistry , Polyketides/pharmacology , Protein Biosynthesis
4.
ACS Synth Biol ; 10(11): 2772-2783, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34677942

ABSTRACT

The process of optimizing the properties of biological molecules is paramount for many industrial and medical applications. Directed evolution is a powerful technique for modifying and improving biomolecules such as proteins or nucleic acids (DNA or RNA). Mimicking the mechanism of natural evolution, one can enhance a desired property by applying a suitable selection pressure and sorting improved variants. Droplet-based microfluidic systems offer a high-throughput solution to this approach by helping to overcome the limiting screening steps and allowing the analysis of variants within increasingly complex libraries. Here, we review cases where successful evolution of biomolecules was achieved using droplet-based microfluidics, focusing on the molecular processes involved and the incorporation of microfluidics to the workflow. We highlight the advantages and limitations of these microfluidic systems compared to low-throughput methods and show how the integration of these systems into directed evolution workflows can open new avenues to discover or improve biomolecules according to user-defined conditions.


Subject(s)
Directed Molecular Evolution/methods , Animals , DNA/genetics , High-Throughput Screening Assays/methods , Humans , Microfluidic Analytical Techniques/methods , Microfluidics/methods , RNA/genetics
5.
Nat Commun ; 12(1): 5340, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504068

ABSTRACT

Free L-tryptophan (L-Trp) stalls ribosomes engaged in the synthesis of TnaC, a leader peptide controlling the expression of the Escherichia coli tryptophanase operon. Despite extensive characterization, the molecular mechanism underlying the recognition and response to L-Trp by the TnaC-ribosome complex remains unknown. Here, we use a combined biochemical and structural approach to characterize a TnaC variant (R23F) with greatly enhanced sensitivity for L-Trp. We show that the TnaC-ribosome complex captures a single L-Trp molecule to undergo termination arrest and that nascent TnaC prevents the catalytic GGQ loop of release factor 2 from adopting an active conformation at the peptidyl transferase center. Importantly, the L-Trp binding site is not altered by the R23F mutation, suggesting that the relative rates of L-Trp binding and peptidyl-tRNA cleavage determine the tryptophan sensitivity of each variant. Thus, our study reveals a strategy whereby a nascent peptide assists the ribosome in detecting a small metabolite.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Peptide Chain Initiation, Translational , Ribosomes/genetics , Tryptophan/chemistry , Amino Acid Substitution , Binding Sites , Cryoelectron Microscopy , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Mutation , Operon , Peptide Chain Termination, Translational , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/metabolism , Ribosomes/ultrastructure , Tryptophan/metabolism
6.
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
7.
FEMS Microbiol Rev ; 44(6): 793-803, 2020 11 24.
Article in English | MEDLINE | ID: mdl-32717057

ABSTRACT

Resistance to antimicrobial drugs used to treat bacterial, viral, fungal and parasitic infections is a major health concern requiring a coordinated response across the globe. An important aspect in the fight against antimicrobial resistance is the development of novel drugs that are effective against resistant pathogens. Drug development is a complex trans-disciplinary endeavor, in which structural biology plays a major role by providing detailed functional and mechanistic information on an antimicrobial target and its interactions with small molecule inhibitors. Although X-ray crystallography and nuclear magnetic resonance have until now been the methods of choice to characterize microbial targets and drive structure-based drug development, cryo-electron microscopy is rapidly gaining ground in these areas. In this perspective, we will discuss how cryo-electron microscopy is changing our understanding of an established antimicrobial target, the ribosome, and how methodological developments could help this technique become an integral part of the antimicrobial drug discovery pipeline.


Subject(s)
Anti-Infective Agents/chemistry , Cryoelectron Microscopy , Drug Development , Ribosomes/metabolism , Drug Resistance, Bacterial
8.
Front Pharmacol ; 11: 532, 2020.
Article in English | MEDLINE | ID: mdl-32390853

ABSTRACT

In the search for new antibiotics to combat multidrug-resistant microbes, insects offer a rich source of novel anti-infectives, including a remarkably diverse array of antimicrobial peptides (AMPs) with broad activity against a wide range of species. Larvae of the common green bottle fly Lucilia sericata are used for maggot debridement therapy, and their effectiveness in part reflects the large panel of AMPs they secrete into the wound. To investigate the activity of these peptides in more detail, we selected two structurally different proline rich peptides (Lser-PRP2 and Lser-PRP3) in addition to the α-helical peptide Lser-stomoxyn. We investigated the mechanism of anti-Escherichia coli action of the PRPs in vitro and found that neither of them interfered with protein synthesis but both were able to bind the bacterial chaperone DnaK and are therefore likely to inhibit protein folding. However, unlike Lser-stomoxyn that permeabilized the bacterial membrane by 1% at the low concentration (0.25 µM) neither of the PRPs alone was able to permeabilize E. coli membrane. In the presence of this Lser-stomoxyn concentration significant increase in anti-E. coli activity of Lser-PRP2 was observed, indicating that this peptide needs specific membrane permeabilizing agents to exert its antibacterial activity. We then examined the AMPs-treated bacterial surface and observed detrimental structural changes in the bacterial cell envelope in response to combined AMPs. The functional analysis of insect AMPs will help select optimal combinations for targeted antimicrobial therapy.

9.
Nat Microbiol ; 5(4): 653-654, 2020 04.
Article in English | MEDLINE | ID: mdl-32218511

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
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
11.
Integr Org Biol ; 2(1): obaa012, 2020.
Article in English | MEDLINE | ID: mdl-33793689

ABSTRACT

Many juvenile Kemp's ridley (Lepidochelys kempii) and loggerhead (Caretta caretta) turtles strand during fall on the beaches of Cape Cod (MA, USA), with total stranding numbers sometimes exceeding 300 turtles per year. Once rehabilitated, turtles must be released at beaches with appropriate water temperatures, often requiring transportation to southeastern coastal states of the USA. These transportation events (transports) may approach or exceed 24 h in duration. Kemp's ridley turtles are known to exhibit an adrenal stress response during such transports, but the effect of transport duration has been unclear, and no other sea turtle species has been investigated. To assess whether transport duration and/or species affects physiological reactions to transport, we studied pre- and post-transport physiological measures in Kemp's ridley and loggerhead turtles transported by ground for <6, ∼12, ∼18, or ∼24 h, comparing with matched "control events" in which turtles were studied without transport. Blood samples were analyzed for four stress-associated measures (corticosterone, glucose, total white blood cell [WBC] count, and heterophil/lymphocyte ratio [H/L]) and nine measures of clinical status (pH, pO2, pCO2, HCO3, sodium, potassium, ionized calcium, lactate, and hematocrit). In both species, stress-associated measures elevated significantly during transport, while handling without transport had no significant effects. Loggerheads exhibited a greater stress response than Kemp's ridleys across all transport durations. These results indicate that sea turtles do react physiologically to ground transport; therefore, minimizing transport time and streamlining transport logistics (where feasible) may help ensure release of rehabilitated turtles to sea in the best possible condition. Nonetheless, both species remained in good clinical condition even after 24 h transport, indicating that current transport protocols are generally safe for sea turtles from a clinical perspective.

12.
Biochemistry ; 58(2): 75-84, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30372045

ABSTRACT

The threat of bacteria resistant to multiple antibiotics poses a major public health problem requiring immediate and coordinated action worldwide. While infectious pathogens have become increasingly resistant to commercially available drugs, antibiotic discovery programs in major pharmaceutical companies have produced no new antibiotic scaffolds in 40 years. As a result, new strategies must be sought to obtain a steady supply of novel scaffolds capable of countering the spread of resistance. The bacterial ribosome is a major target for antimicrobials and is inhibited by more than half of the antibiotics used today. Recent studies showing that the ribosome is a target for several classes of ribosomally synthesized antimicrobial peptides point to ribosome-targeting peptides as a promising source of antibiotic scaffolds. In this Perspective, we revisit the current paradigm of antibiotic discovery by proposing that the bacterial ribosome can be used both as a target and as a tool for the production and selection of peptide-based antimicrobials. Turning the ribosome into a high-throughput platform for the directed evolution of peptide-based antibiotics could be achieved in different ways. One possibility would be to use a combination of state-of-the-art microfluidics and genetic reprogramming techniques, which we will review briefly. If it is successful, this strategy has the potential to produce new classes of antibiotics for treating multi-drug-resistant pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Peptides/metabolism , Peptides/pharmacology , Ribosomes/drug effects , Anti-Bacterial Agents/chemistry , Bacteria/genetics , Bacteria/metabolism , Directed Molecular Evolution/methods , Drug Discovery , Drug Evaluation, Preclinical/methods , Genetic Association Studies , High-Throughput Screening Assays/methods , Peptide Library , Peptides/chemistry , Protein Biosynthesis/drug effects , Ribosomes/genetics , Ribosomes/metabolism
13.
Life Sci Alliance ; 1(5): e201800148, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30456383

ABSTRACT

Although it is known that the amino acid sequence of a nascent polypeptide can impact its rate of translation, dedicated tools to systematically investigate this process are lacking. Here, we present high-throughput inverse toeprinting, a method to identify peptide-encoding transcripts that induce ribosomal stalling in vitro. Unlike ribosome profiling, inverse toeprinting protects the entire coding region upstream of a stalled ribosome, making it possible to work with random or focused transcript libraries that efficiently sample the sequence space. We used inverse toeprinting to characterize the stalling landscapes of free and drug-bound Escherichia coli ribosomes, obtaining a comprehensive list of arrest motifs that were validated in vivo, along with a quantitative measure of their pause strength. Thanks to the modest sequencing depth and small amounts of material required, inverse toeprinting provides a highly scalable and versatile tool to study sequence-dependent translational processes.

14.
Cell Chem Biol ; 25(5): 530-539.e7, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29526712

ABSTRACT

Proline-rich antimicrobial peptides (PrAMPs) internalize into susceptible bacteria using specific transporters and interfere with protein synthesis and folding. To date, mammalian PrAMPs have so far been identified only in artiodactyls. Since cetaceans are co-phyletic with artiodactyls, we mined the genome of the bottlenose dolphin Tursiops truncatus, leading to the identification of two PrAMPs, Tur1A and Tur1B. Tur1A, which is orthologous to the bovine PrAMP Bac7, is internalized into Escherichia coli, without damaging the membranes, using the inner membrane transporters SbmA and YjiL/MdM. Furthermore, like Bac7, Tur1A also inhibits bacterial protein synthesis by binding to the ribosome and blocking the transition from the initiation to the elongation phase. By contrast, Tur1B is a poor inhibitor of protein synthesis and may utilize another mechanism of action. An X-ray structure of Tur1A bound within the ribosomal exit tunnel provides a basis to develop these peptides as novel antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Escherichia coli/drug effects , Protein Biosynthesis/drug effects , Ribosomes/drug effects , Animals , Crystallography, X-Ray , Dolphins , Escherichia coli/metabolism , Escherichia coli Infections/drug therapy , Escherichia coli Proteins/metabolism , Humans , Models, Molecular , Ribosomes/metabolism
15.
Nat Prod Rep ; 34(7): 702-711, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28537612

ABSTRACT

Covering: up to 2017The innate immune system employs a broad array of antimicrobial peptides (AMPs) to attack invading microorganisms. While most AMPs act by permeabilizing the bacterial membrane, specific subclasses of AMPs have been identified that pass through membranes and inhibit bacterial growth by targeting fundamental intracellular processes. One such subclass is the proline-rich antimicrobial peptides (PrAMPs) that bind to the ribosome and interfere with the process of protein synthesis. A diverse range of PrAMPs have been identified in insects, such as bees, wasps and beetles, and crustaceans, such as crabs, as well as in mammals, such as cows, sheep, goats and pigs. Mechanistically, the best-characterized PrAMPs are the insect oncocins, such as Onc112, and bovine bactenecins, such as Bac7. Biochemical and structural studies have revealed that these PrAMPs bind within the ribosomal exit tunnel with a reverse orientation compared to a nascent polypeptide chain. The PrAMPs allow initiation but prevent the transition into the elongation phase of translation. Insight into the interactions of PrAMPs with their ribosomal target provides the opportunity to further develop these peptides as novel antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Infective Agents/chemical synthesis , Antimicrobial Cationic Peptides/chemical synthesis , Proline/chemistry , Animals , Anti-Bacterial Agents/chemistry , Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Cattle , Coleoptera , Female , Microbial Sensitivity Tests , Peptides/metabolism , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Sheep , Swine , Wasps
16.
Nat Commun ; 7: 12026, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27380950

ABSTRACT

Nascent polypeptides can induce ribosome stalling, regulating downstream genes. Stalling of ErmBL peptide translation in the presence of the macrolide antibiotic erythromycin leads to resistance in Streptococcus sanguis. To reveal this stalling mechanism we obtained 3.6-Å-resolution cryo-EM structures of ErmBL-stalled ribosomes with erythromycin. The nascent peptide adopts an unusual conformation with the C-terminal Asp10 side chain in a previously unseen rotated position. Together with molecular dynamics simulations, the structures indicate that peptide-bond formation is inhibited by displacement of the peptidyl-tRNA A76 ribose from its canonical position, and by non-productive interactions of the A-tRNA Lys11 side chain with the A-site crevice. These two effects combine to perturb peptide-bond formation by increasing the distance between the attacking Lys11 amine and the Asp10 carbonyl carbon. The interplay between drug, peptide and ribosome uncovered here also provides insight into the fundamental mechanism of peptide-bond formation.


Subject(s)
Bacterial Proteins/chemistry , Protein Biosynthesis/drug effects , RNA, Transfer, Amino Acyl/chemistry , Ribosomes/metabolism , Streptococcus sanguis/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cryoelectron Microscopy , Erythromycin/chemistry , Erythromycin/pharmacology , Internal Ribosome Entry Sites , Molecular Dynamics Simulation , Nucleic Acid Conformation , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/drug effects , Ribosomes/ultrastructure , Streptococcus sanguis/drug effects , Streptococcus sanguis/metabolism
17.
J Mol Biol ; 428(10 Pt B): 2217-27, 2016 05 22.
Article in English | MEDLINE | ID: mdl-27108680

ABSTRACT

In order to colonize a niche and compete for scarce resources, microorganisms have evolved means to adjust the expression levels of their biosynthetic enzymes in response to the changing levels of metabolites available to them. To do so, they often rely on transcription factors or structured RNAs that directly sense the concentration of metabolites and turn genes on or off accordingly. In some instances, however, a metabolite can be sensed by an actively translating ribosome bearing a nascent polypeptide whose specific amino acid sequence interferes with translation. These "arrest peptides" lead to the formation of stalled ribosome nascent chain complexes on the mRNA that can regulate the expression of downstream genes through transcriptional or translational mechanisms. Although this process was discovered over three and a half decades ago, the extent to which arrest peptides regulate gene expression in response to cell metabolites is unknown. Here, we examine the physical constraints imposed by the ribosome on peptide-mediated ligand sensing and review attempts to assess the diversity of arrest peptides to date. In addition, we outline a possible way forward to establish how pervasive metabolite sensing by arrest peptides is in nature.


Subject(s)
Gene Expression Regulation, Bacterial , Peptide Biosynthesis , Protein Biosynthesis/drug effects , Ribosomes/drug effects , Ribosomes/metabolism , Models, Biological , Ribosomes/chemistry
18.
Nucleic Acids Res ; 44(5): 2429-38, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26792896

ABSTRACT

Proline-rich antimicrobial peptides (PrAMPs) produced as part of the innate immune response of animals, insects and plants represent a vast, untapped resource for the treatment of multidrug-resistant bacterial infections. PrAMPs such as oncocin or bactenecin-7 (Bac7) interact with the bacterial ribosome to inhibit translation, but their supposed specificity as inhibitors of bacterial rather than mammalian protein synthesis remains unclear, despite being key to developing drugs with low toxicity. Here, we present crystal structures of the Thermus thermophilus 70S ribosome in complex with the first 16 residues of mammalian Bac7, as well as the insect-derived PrAMPs metalnikowin I and pyrrhocoricin. The structures reveal that the mammalian Bac7 interacts with a similar region of the ribosome as insect-derived PrAMPs. Consistently, Bac7 and the oncocin derivative Onc112 compete effectively with antibiotics, such as erythromycin, which target the ribosomal exit tunnel. Moreover, we demonstrate that Bac7 allows initiation complex formation but prevents entry into the elongation phase of translation, and show that it inhibits translation on both mammalian and bacterial ribosomes, explaining why this peptide needs to be stored as an inactive pro-peptide. These findings highlight the need to consider the specificity of PrAMP derivatives for the bacterial ribosome in future drug development efforts.


Subject(s)
Anti-Bacterial Agents/chemistry , Peptides, Cyclic/chemistry , Protein Biosynthesis/drug effects , Ribosomes/drug effects , Amino Acid Sequence , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Binding Sites , Binding, Competitive , Cattle , Crystallography, X-Ray , Erythromycin/chemistry , Erythromycin/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Heteroptera/chemistry , Insect Proteins/chemistry , Insect Proteins/pharmacology , Models, Molecular , Molecular Sequence Data , Peptides, Cyclic/pharmacology , Protein Binding , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Species Specificity , Thermus thermophilus/chemistry
20.
Nat Struct Mol Biol ; 22(6): 470-5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25984971

ABSTRACT

The increasing prevalence of multidrug-resistant pathogenic bacteria is making current antibiotics obsolete. Proline-rich antimicrobial peptides (PrAMPs) display potent activity against Gram-negative bacteria and thus represent an avenue for antibiotic development. PrAMPs from the oncocin family interact with the ribosome to inhibit translation, but their mode of action has remained unclear. Here we have determined a structure of the Onc112 peptide in complex with the Thermus thermophilus 70S ribosome at a resolution of 3.1 Å by X-ray crystallography. The Onc112 peptide binds within the ribosomal exit tunnel and extends toward the peptidyl transferase center, where it overlaps with the binding site for an aminoacyl-tRNA. We show biochemically that the binding of Onc112 blocks and destabilizes the initiation complex, thus preventing entry into the elongation phase. Our findings provide a basis for the future development of this class of potent antimicrobial agents.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Peptide Chain Initiation, Translational/drug effects , Protein Synthesis Inhibitors/pharmacology , Ribosomes/chemistry , Antimicrobial Cationic Peptides/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Protein Synthesis Inhibitors/metabolism , Ribosomes/metabolism , Thermus thermophilus/chemistry , Thermus thermophilus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...