Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Org Lett ; 26(21): 4433-4437, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38767867

ABSTRACT

Most biosynthetic gene clusters (BGCs) are functionally inaccessible by using fermentation methods. Bioinformatic-coupled total synthesis provides an alternative approach for accessing BGC-encoded bioactivities. To date, synthetic bioinformatic natural product (synBNP) methods have focused on lipopeptides containing simple lipids. Here we increase the bioinformatic and synthetic complexity of the synBNP approach by targeting BGCs that encode N-cinnamoyl lipids. This led to our synthesis of cinnamosyn, a 10-mer N-cinnamoyl-containing peptide that is cytotoxic to human cells.


Subject(s)
Biological Products , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/chemical synthesis , Humans , Molecular Structure , Computational Biology , Multigene Family , Lipopeptides/chemistry , Lipopeptides/pharmacology , Lipopeptides/chemical synthesis , Cinnamates/chemistry , Cinnamates/pharmacology , Cinnamates/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis
2.
Org Lett ; 24(27): 4943-4948, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35776528

ABSTRACT

Cationic nonribosomal lipopeptides (CNRLPs) from Paenibacillus spp. have been a rewarding source of Gram-negative-active antibiotics. Here we systematically screened sequenced bacterial genomes for CNRLP biosynthetic gene clusters (BGCs) that we predicted might encode additional Gram-negative-active antibiotics. Total chemical synthesis of the bioinformatically predicted products of seven such BGCs led to our identification of new laterocidine, tridecaptin, and paenibacterin-like antibiotics with potent activity against both multiple-drug-resistant Gram-negative and Gram-positive pathogens.


Subject(s)
Anti-Bacterial Agents , Paenibacillus , Anti-Bacterial Agents/pharmacology , Genome, Bacterial , Lipopeptides/pharmacology , Multigene Family , Paenibacillus/genetics
3.
Science ; 376(6596): 991-996, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35617397

ABSTRACT

Emerging resistance to currently used antibiotics is a global public health crisis. Because most of the biosynthetic capacity within the bacterial kingdom has remained silent in previous antibiotic discovery efforts, uncharacterized biosynthetic gene clusters found in bacterial genome-sequencing studies remain an appealing source of antibiotics with distinctive modes of action. Here, we report the discovery of a naturally inspired lipopeptide antibiotic called cilagicin, which we chemically synthesized on the basis of a detailed bioinformatic analysis of the cil biosynthetic gene cluster. Cilagicin's ability to sequester two distinct, indispensable undecaprenyl phosphates used in cell wall biosynthesis, together with the absence of detectable resistance in laboratory tests and among multidrug-resistant clinical isolates, makes it an appealing candidate for combating antibiotic-resistant pathogens.


Subject(s)
Anti-Bacterial Agents , Lipopeptides , Anti-Bacterial Agents/pharmacology , Computational Biology , Lipopeptides/pharmacology , Microbial Sensitivity Tests , Multigene Family
4.
Nature ; 601(7894): 606-611, 2022 01.
Article in English | MEDLINE | ID: mdl-34987225

ABSTRACT

Gram-negative bacteria are responsible for an increasing number of deaths caused by antibiotic-resistant infections1,2. The bacterial natural product colistin is considered the last line of defence against a number of Gram-negative pathogens. The recent global spread of the plasmid-borne mobilized colistin-resistance gene mcr-1 (phosphoethanolamine transferase) threatens the usefulness of colistin3. Bacteria-derived antibiotics often appear in nature as collections of similar structures that are encoded by evolutionarily related biosynthetic gene clusters. This structural diversity is, at least in part, expected to be a response to the development of natural resistance, which often mechanistically mimics clinical resistance. Here we propose that a solution to mcr-1-mediated resistance might have evolved among naturally occurring colistin congeners. Bioinformatic analysis of sequenced bacterial genomes identified a biosynthetic gene cluster that was predicted to encode a structurally divergent colistin congener. Chemical synthesis of this structure produced macolacin, which is active against Gram-negative pathogens expressing mcr-1 and intrinsically resistant pathogens with chromosomally encoded phosphoethanolamine transferase genes. These Gram-negative bacteria include extensively drug-resistant Acinetobacter baumannii and intrinsically colistin-resistant Neisseria gonorrhoeae, which, owing to a lack of effective treatment options, are considered among the highest level threat pathogens4. In a mouse neutropenic infection model, a biphenyl analogue of macolacin proved to be effective against extensively drug-resistant A. baumannii with colistin-resistance, thus providing a naturally inspired and easily produced therapeutic lead for overcoming colistin-resistant pathogens.


Subject(s)
Anti-Bacterial Agents , Colistin , Drug Resistance, Bacterial , Gram-Negative Bacteria , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/enzymology , Acinetobacter baumannii/genetics , Animals , Anti-Bacterial Agents/pharmacology , Biosynthetic Pathways/genetics , Colistin/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Ethanolamines , Genes, Bacterial , Genome, Bacterial , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Gram-Negative Bacteria/genetics , Mice , Microbial Sensitivity Tests , Multigene Family , Neutropenia/drug therapy , Neutropenia/microbiology , Plasmids , Transferases (Other Substituted Phosphate Groups)
5.
Bioorg Med Chem Lett ; 57: 128484, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34861348

ABSTRACT

Bacterial resistance threatens the utility of currently available antibiotics. Rifampicin, a cornerstone in the treatment of persistent Gram-positive infections, is prone to the development of resistance resulting from single point mutations in the antibiotic's target, RNA polymerase. One strategy to circumvent resistance is the use of 'hybrid' antibiotics consisting of two covalently linked antibiotic entities. These compounds generally have two distinct cellular targets, reducing the probability of resistance development and potentially providing simplified pharmacological properties compared to combination therapies using the individual antibiotics. Here we evaluate a series of semi-synthetic hybrid antibiotics formed by linking kanglemycin A (Kang A), a rifampicin analog, and a collection of fluoroquinolones. Kang A is a natural product antibiotic which contains a novel dimethyl succinic acid moiety that offers a new attachment point for the synthesis of hybrid antibiotics. We compare the activity of the Kang A hybrids generated via the acid attachment point to a series of hybrids linked at the compound's naphthoquinone ring system. Several hybrids exhibit activity against bacteria resistant to Kang A via the action of the partnered antibiotic, suggesting that the Kang scaffold may provide new avenues for generating antibiotics effective against drug-resistant infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fluoroquinolones/pharmacology , Rifamycins/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Drug Resistance, Bacterial/drug effects , Escherichia coli/drug effects , Fluoroquinolones/chemical synthesis , Fluoroquinolones/toxicity , HEK293 Cells , Humans , Microbial Sensitivity Tests , Rifamycins/chemical synthesis , Rifamycins/toxicity , Staphylococcus aureus/drug effects
6.
Nat Microbiol ; 7(1): 120-131, 2022 01.
Article in English | MEDLINE | ID: mdl-34949828

ABSTRACT

The emergence of multidrug-resistant bacteria poses a threat to global health and necessitates the development of additional in vivo active antibiotics with diverse modes of action. Directly targeting menaquinone (MK), which plays an important role in bacterial electron transport, is an appealing, yet underexplored, mode of action due to a dearth of MK-binding molecules. Here we combine sequence-based metagenomic mining with a motif search of bioinformatically predicted natural product structures to identify six biosynthetic gene clusters that we predicted encode MK-binding antibiotics (MBAs). Their predicted products (MBA1-6) were rapidly accessed using a synthetic bioinformatic natural product approach, which relies on bioinformatic structure prediction followed by chemical synthesis. Among these six structurally diverse MBAs, four make up two new MBA structural families. The most potent member of each new family (MBA3, MBA6) proved effective at treating methicillin-resistant Staphylococcus aureus infection in a murine peritonitis-sepsis model. The only conserved feature present in all MBAs is the sequence 'GXLXXXW', which we propose represents a minimum MK-binding motif. Notably, we found that a subset of MBAs were active against Mycobacterium tuberculosis both in vitro and in macrophages. Our findings suggest that naturally occurring MBAs are a structurally diverse and untapped class of mechanistically interesting, in vivo active antibiotics.


Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Methicillin-Resistant Staphylococcus aureus/drug effects , Vitamin K 2/metabolism , Animals , Drug Resistance, Multiple, Bacterial , Female , Humans , Macrophages/drug effects , Macrophages/microbiology , Metagenomics/methods , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Peritonitis/drug therapy , Peritonitis/microbiology , Staphylococcal Infections/drug therapy , Vitamin K 2/isolation & purification
7.
mSphere ; 6(6): e0071121, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34851166

ABSTRACT

The COVID-19 pandemic has highlighted the need to identify additional antiviral small molecules to complement existing therapies. Although increasing evidence suggests that metabolites produced by the human microbiome have diverse biological activities, their antiviral properties remain poorly explored. Using a cell-based SARS-CoV-2 infection assay, we screened culture broth extracts from a collection of phylogenetically diverse human-associated bacteria for the production of small molecules with antiviral activity. Bioassay-guided fractionation uncovered three bacterial metabolites capable of inhibiting SARS-CoV-2 infection. This included the nucleoside analogue N6-(Δ2-isopentenyl)adenosine, the 5-hydroxytryptamine receptor agonist tryptamine, and the pyrazine 2,5-bis(3-indolylmethyl)pyrazine. The most potent of these, N6-(Δ2-isopentenyl)adenosine, had a 50% inhibitory concentration (IC50) of 2 µM. These natural antiviral compounds exhibit structural and functional similarities to synthetic drugs that have been clinically examined for use against COVID-19. Our discovery of structurally diverse metabolites with anti-SARS-CoV-2 activity from screening a small fraction of the bacteria reported to be associated with the human microbiome suggests that continued exploration of phylogenetically diverse human-associated bacteria is likely to uncover additional small molecules that inhibit SARS-CoV-2 as well as other viral infections. IMPORTANCE The continued prevalence of COVID-19 and the emergence of new variants has once again put the spotlight on the need for the identification of SARS-CoV-2 antivirals. The human microbiome produces an array of small molecules with bioactivities (e.g., host receptor ligands), but its ability to produce antiviral small molecules is relatively underexplored. Here, using a cell-based screening platform, we describe the isolation of three microbiome-derived metabolites that are able to prevent SARS-CoV-2 infection in vitro. These molecules display structural similarities to synthetic drugs that have been explored for the treatment of COVID-19, and these results suggest that the microbiome may be a fruitful source of the discovery of small molecules with antiviral activities.


Subject(s)
Antiviral Agents/pharmacology , Bacteria/metabolism , Culture Media/chemistry , Metabolic Networks and Pathways , Microbiota/physiology , SARS-CoV-2/drug effects , Symbiosis/physiology , Bacteria/chemistry , Bacteria/classification , Bacteria/growth & development , Biological Assay , Cell Line, Tumor , Culture Media/pharmacology , Humans , Molecular Docking Simulation , Protease Inhibitors/pharmacology , Protein Binding
8.
Chembiochem ; 22(11): 1940-1947, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33644965

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is a human pathobiont that causes drastic antibiotic-resistant infections and is responsible for millions of deaths universally. Pneumococcus pathogenicity relies on the competence-stimulating peptide (CSP)-mediated quorum-sensing (QS) pathway that controls competence development for genetic transformation and, consequently, the spread of antibiotic resistance and virulence genes. Modulation of QS in S. pneumoniae can therefore be used to enervate pneumococcal infectivity as well as minimize the susceptibility to resistance development. In this work, we sought to optimize the interaction of CSP1 with its cognate transmembrane histidine kinase receptor (ComD1) through substitution of proteogenic and nonproteogenic amino acids on the hydrophobic binding face of CSP1. The findings from this study not only provided additional structure-activity data that are significant in optimizing CSP1 potency, but also led to the development of potent QS modulators. These CSP-based QS modulators could be used as privileged scaffolds for the development of antimicrobial agents against pneumococcal infections.


Subject(s)
Peptides/metabolism , Streptococcus pneumoniae/metabolism , Hydrophobic and Hydrophilic Interactions , Peptides/chemistry , Quorum Sensing , Streptococcus pneumoniae/chemistry
9.
J Nat Prod ; 84(4): 1056-1066, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33621083

ABSTRACT

Tuberculosis (TB) remains one of the deadliest infectious diseases. Unfortunately, the development of antibiotic resistance threatens our current therapeutic arsenal, which has necessitated the discovery and development of novel antibiotics against drug-resistant Mycobacterium tuberculosis (Mtb). Cyclomarin A and rufomycin I are structurally related cyclic heptapeptides assembled by nonribosomal peptide synthetases (NRPSs), which show potent anti-Mtb activity with a new cellular target, the caseinolytic protein ClpC1. An NRPS adenylation domain survey using DNA extracted from ∼2000 ecologically diverse soils found low cyclomarin/rufomycin biosynthetic diversity. In this survey, a family of cyclomarin/rufomycin-like biosynthetic gene clusters (BGC) that encode metamarin, an uncommon cyclomarin congener with potent activity against both Mtb H37Rv and multidrug-resistant Mtb clinical isolates was identified. Metamarin effectively inhibits Mtb growth in murine macrophages and increases the activities of ClpC1 ATPase and the associated ClpC1/P1/P2 protease complex, thus causing cell death by uncontrolled protein degradation.


Subject(s)
Metagenome , Mycobacterium tuberculosis/drug effects , Oligopeptides/pharmacology , Soil Microbiology , Animals , Antitubercular Agents , Bacterial Proteins , Cell Line , Heat-Shock Proteins , Macrophages , Mice , Microbial Sensitivity Tests , Molecular Structure
10.
J Am Chem Soc ; 142(33): 14158-14168, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32697091

ABSTRACT

Bacterial natural products have inspired the development of numerous antibiotics in use today. As resistance to existing antibiotics has become more prevalent, new antibiotic lead structures and activities are desperately needed. An increasing number of natural product biosynthetic gene clusters, to which no known molecules can be assigned, are found in genome and metagenome sequencing data. Here we access structural information encoded in this underexploited resource using a synthetic-bioinformatic natural product (syn-BNP) approach, which relies on bioinformatic algorithms followed by chemical synthesis to predict and then produce small molecules inspired by biosynthetic gene clusters. In total, 157 syn-BNP cyclic peptides inspired by 96 nonribosomal peptide synthetase gene clusters were synthesized and screened for antibacterial activity. This yielded nine antibiotics with activities against ESKAPE pathogens as well as Mycobacterium tuberculosis. Not only are antibiotic-resistant pathogens susceptible to many of these syn-BNP antibiotics, but they were also unable to develop resistance to these antibiotics in laboratory experiments. Characterized modes of action for these antibiotics include cell lysis, membrane depolarization, inhibition of cell wall biosynthesis, and ClpP protease dysregulation. Increasingly refined syn-BNP-based explorations of biosynthetic gene clusters should allow for more rapid identification of evolutionarily inspired bioactive small molecules, in particular antibiotics with diverse mechanism of actions that could help confront the imminent crisis of antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biological Products/pharmacology , Computational Biology , Mycobacterium tuberculosis/drug effects , Algorithms , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Microbial Sensitivity Tests , Molecular Structure
11.
Chembiochem ; 21(3): 340-345, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31291510

ABSTRACT

The rapid increase in multidrug-resistant pathogens is a major health concern that could bring mankind back to the pre-antibiotic era. Streptococcus pneumoniae is a highly recombinogenic opportunistic pathogen that causes a variety of deadly diseases and rapidly develops resistance to current antibiotic treatments. S. pneumoniae pathogenicity is dependent on a cell-density communication mechanism, or quorum sensing (QS), termed the competence regulon. In this work, we set out to design signal-based QS modulators capable of affecting the two specificity groups found in S. pneumoniae. Through systematic analysis and rational design, we were able to construct peptide-based pan-group QS activators and inhibitors with activities in the nanomolar range. These novel analogues are privileged scaffolds for the development of anti-virulence therapeutics against S. pneumoniae infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Peptides/pharmacology , Pneumococcal Infections/drug therapy , Streptococcus pneumoniae/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Peptides/chemical synthesis , Peptides/chemistry , Quorum Sensing/drug effects , Solid-Phase Synthesis Techniques
12.
ACS Med Chem Lett ; 10(6): 880-886, 2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31223442

ABSTRACT

Streptococcus pneumoniae is an opportunistic pathogen that can cause diseases ranging from mild respiratory infections to life-threatening conditions such as pneumonia, meningitis, and bacteremia. S. pneumoniae pathogenicity is dependent on the action of a 17-amino acid peptide pheromone, termed competence stimulating peptide (CSP) that controls the competence regulon, a quorum sensing (QS) circuit. Therefore, intercepting QS could have therapeutic implications in treating pneumococcal infections while avoiding emerging antimicrobial resistance. In this study, we set out to evaluate the impact of amide protons on CSP activity and metabolic stability through systematic N-methylation. Our results indicate that the majority of amide protons are critical for CSP activity, either through direct interactions with the cognate receptor or by stabilizing the bioactive conformation. Importantly, we identified several N-methyl CSP analogs, namely, CSP1(15)-N-Me-K6 and CSP1(15)-N-Me-F7, that retain their biological activity while exhibiting enhanced metabolic stability. These analogs are privileged scaffolds for the design of CSP-based QS modulators with drug-like properties.

13.
Chembiochem ; 19(22): 2380-2386, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30211457

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is a prevalent human pathogen responsible for a variety of diseases, including pneumonia, bacteremia, sepsis, meningitis and otitis media, with a death toll of >22 000 a year in the United States alone. Pneumococcus uses the competence regulon and its associated signaling peptide, the competence stimulating peptide (CSP), to initiate its attack on the host and establish an infection. In this work, we set out to: 1) develop a pan-group quorum sensing inhibitor that could effectively interact with both the pneumococcus ComD1 and ComD2 receptors; and 2) evaluate the utility of dominant-negative CSPs (dnCSPs) in attenuating pneumococcus infectivity. Our results highlight the potential of inhibiting the competence regulon as a therapeutic approach to combat pneumococcus infections.


Subject(s)
Bacterial Proteins , Pneumonia, Pneumococcal , Quorum Sensing/drug effects , Streptococcus pneumoniae , Acute Disease , Animals , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , Disease Models, Animal , Humans , Mice , Molecular Targeted Therapy , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Virulence
14.
Beilstein J Org Chem ; 14: 1769-1777, 2018.
Article in English | MEDLINE | ID: mdl-30112082

ABSTRACT

Quorum sensing (QS) is a cell-cell communication mechanism that enables bacteria to assess their population density and alter their behavior upon reaching high cell number. Many bacterial pathogens utilize QS to initiate an attack on their host, thus QS has attracted significant attention as a potential antivirulence alternative to traditional antibiotics. Streptococcus pneumoniae, a notorious human pathogen responsible for a variety of acute and chronic infections, utilizes the competence regulon and its associated signaling peptide, the competence stimulating peptide (CSP), to acquire antibiotic resistance and establish an infection. In this work, we sought to define the binding pockets within the ComD1 receptor used for binding the hydrophobic side-chain residues in CSP1 through the introduction of highly-conservative point mutations within the peptide. Optimization of these binding interactions could lead to the development of highly potent CSP-based QS modulators while the inclusion of non-natural amino acids within the CSP sequence would confer resistance to protease degradation, a requirement for drug candidates.

15.
ACS Chem Biol ; 12(4): 1141-1151, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28221753

ABSTRACT

Streptococcus pneumoniae is a highly recombinogenic human pathogen that utilizes the competence stimulating peptide (CSP)-based quorum sensing (QS) circuitry to acquire antibiotic resistance genes from the environment and initiate its attack on the human host. Modulation of QS in this bacterium, either inhibition or activation, can therefore be used to attenuate S. pneumoniae infectivity and slow down pneumococcal resistance development. In this study, we set to determine the molecular mechanism that drives CSP:receptor binding and identify CSP-based QS modulators with distinct activity profiles. To this end, we conducted systematic replacement of the amino acid residues in the two major CSP signals (CSP1 and CSP2) and assessed the ability of the mutated analogs to modulate QS against both cognate and noncognate ComD receptors. We then evaluated the overall 3D structures of these analogs using circular dichroism (CD) to correlate between the structure and function of these peptides. Our CD analysis revealed a strong correlation between α-helicity and bioactivity for both specificity groups (CSP1 and CSP2). Furthermore, we identified the first pan-group QS activator and the most potent group-II QS inhibitor to date. These chemical probes can be used to study the role of QS in S. pneumoniae and as scaffolds for the design of QS-based anti-infective therapeutics against S. pneumoniae infections.


Subject(s)
Bacterial Proteins/metabolism , Streptococcus pneumoniae/metabolism , Bacterial Proteins/chemistry , Binding Sites , Circular Dichroism , Genes, Bacterial , Protein Conformation , Quorum Sensing , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/physiology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...