Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Med Chem Lett ; 14(11): 1524-1530, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37974942

ABSTRACT

Multidrug-resistant bacteria are spreading at alarming rates, and despite extensive efforts, no new antibiotic class with activity against Gram-negative bacteria has been approved in over 50 years. LepB inhibitors (LepBi) based on the arylomycin class of natural products are a novel class of antibiotics and function by inhibiting the bacterial type I signal peptidase (SPase) in Gram-negative bacteria. One critical aspect of LepBi development involves optimization of the membrane-anchored lipophilic portion of the molecule. We therefore developed an approach that assesses the effect of this portion on the complicated equilibria of plasma protein binding, crossing the outer membrane of Gram-negative bacteria and anchoring in the bacterial inner membrane to facilitate SPase binding. Our findings provide important insights into the development of antibacterial agents where the target is associated with the inner membrane of Gram-negative bacteria.

2.
Nature ; 561(7722): 189-194, 2018 09.
Article in English | MEDLINE | ID: mdl-30209367

ABSTRACT

Multidrug-resistant bacteria are spreading at alarming rates, and despite extensive efforts no new class of antibiotic with activity against Gram-negative bacteria has been approved in over fifty years. Natural products and their derivatives have a key role in combating Gram-negative pathogens. Here we report chemical optimization of the arylomycins-a class of natural products with weak activity and limited spectrum-to obtain G0775, a molecule with potent, broad-spectrum activity against Gram-negative bacteria. G0775 inhibits the essential bacterial type I signal peptidase, a new antibiotic target, through an unprecedented molecular mechanism. It circumvents existing antibiotic resistance mechanisms and retains activity against contemporary multidrug-resistant Gram-negative clinical isolates in vitro and in several in vivo infection models. These findings demonstrate that optimized arylomycin analogues such as G0775 could translate into new therapies to address the growing threat of multidrug-resistant Gram-negative infections.


Subject(s)
Anti-Bacterial Agents/classification , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Peptides, Cyclic/pharmacology , Biocatalysis/drug effects , Biological Products/classification , Biological Products/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/enzymology , Gram-Negative Bacteria/enzymology , Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/pathogenicity , Lysine/metabolism , Membrane Proteins/antagonists & inhibitors , Microbial Sensitivity Tests , Peptides, Cyclic/chemistry , Porins , Protein Binding , Protein Domains , Serine Endopeptidases , Substrate Specificity
3.
J Med Chem ; 54(14): 4954-63, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21630667

ABSTRACT

While most clinically used antibiotics were derived from natural products, the isolation of new broad-spectrum natural products has become increasingly rare and narrow-spectrum agents are typically deemed unsuitable for development because of intrinsic limitations of their scaffold or target. However, it is possible that the spectrum of a natural product antibiotic might be limited by specific resistance mechanisms in some bacteria, such as target mutations, and the spectra of such "latent" antibiotics might be reoptimized by derivatization, just as has been done with clinically deployed antibiotics. We recently showed that the spectrum of the arylomycin natural product antibiotics, which act via the novel mechanism of inhibiting type I signal peptidase, is broader than previously believed and that resistance in several key human pathogens is due to the presence of a specific Pro residue in the target peptidase that disrupts interactions with the lipopeptide tail of the antibiotic. To begin to test whether this natural resistance might be overcome by derivatization, we synthesized analogues with altered lipopeptide tails and identified several with an increased spectrum of activity against S. aureus. The data support the hypothesis that the arylomycins are latent antibiotics, suggest that their spectrum may be optimized by derivatization, and identify a promising scaffold upon which future optimization efforts might focus.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Lipopeptides/chemical synthesis , Oligopeptides/chemical synthesis , Peptides, Cyclic/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Escherichia coli/drug effects , Lipopeptides/chemistry , Lipopeptides/pharmacology , Membrane Proteins/antagonists & inhibitors , Microbial Sensitivity Tests , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Pseudomonas aeruginosa/drug effects , Serine Endopeptidases , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Structure-Activity Relationship
4.
J Nat Prod ; 74(5): 956-61, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21545107

ABSTRACT

Antibiotics are virtually always isolated as families of related compounds, but the evolutionary forces underlying the observed diversity are generally poorly understood, and it is not even clear whether they are all expected to be biologically active. The arylomycin class of antibiotics is comprised of three related families that are differentiated by nitration, glycosylation, and hydroxylation of a conserved core scaffold. Previously, we reported the total synthesis of an A series member, arylomycin A2, as well as the A series derivative arylomycin C16 and showed that both are active against a broader spectrum of bacteria than previously appreciated. We now report the total synthesis of a B series analogue, arylomycin B-C16, and its aromatic amine derivative. While the aromatic amine loses activity against all bacteria tested, the B series compound shows activities that are similar to the A series compounds, except that it also gains activity against the important pathogen Streptococcus agalactiae.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Microbial Sensitivity Tests , Molecular Structure , Oligopeptides/chemistry , Streptococcus agalactiae/drug effects , Structure-Activity Relationship
5.
Antimicrob Agents Chemother ; 55(3): 1130-4, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21189343

ABSTRACT

The arylomycins are a class of natural-product antibiotics that act via the inhibition of type I signal peptidase (SPase), and we have found in diverse bacteria that their activity is limited by the presence of a resistance-conferring Pro residue in SPase that reduces inhibitor binding. We have also demonstrated that Staphylococcus epidermidis, which lacks this Pro residue, is extremely susceptible to the arylomycins. Here, to further explore the potential utility of the arylomycins, we report an analysis of the activity of a synthetic arylomycin derivative, arylomycin C16, against clinical isolates of S. epidermidis and other coagulase-negative staphylococci (CoNS) from distinct geographical locations. Against many important species of CoNS, including S. epidermidis, S. haemolyticus, S. lugdunensis, and S. hominis, we find that arylomycin C16 exhibits activity equal to or greater than that of vancomycin, the antibiotic most commonly used to treat CoNS infections. While the susceptibility was generally correlated with the absence of the previously identified Pro residue, several cases were identified where additional factors also appear to contribute.


Subject(s)
Anti-Bacterial Agents/pharmacology , Oligopeptides/pharmacology , Staphylococcus epidermidis/drug effects , Staphylococcus/drug effects , Microbial Sensitivity Tests , Molecular Structure , Oligopeptides/chemistry
6.
J Bacteriol ; 193(2): 340-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21075926

ABSTRACT

Bacterial protein secretion is a highly orchestrated process that is essential for infection and virulence. Despite extensive efforts to predict or experimentally detect proteins that are secreted, the characterization of the bacterial secretome has remained challenging. A central event in protein secretion is the type I signal peptidase (SPase)-mediated cleavage of the N-terminal signal peptide that targets a protein for secretion via the general secretory pathway, and the arylomycins are a class of natural products that inhibit SPase, suggesting that they may be useful chemical biology tools for characterizing the secretome. Here, using an arylomycin derivative, along with two-dimensional gel electrophoresis and liquid chromatography-tandem mass spectrometry (LC-MS/MS), we identify 11 proteins whose secretion from stationary-phase Staphylococcus epidermidis is dependent on SPase activity, 9 of which are predicted to be translated with canonical N-terminal signal peptides. In addition, we find that the presence of extracellular domains of lipoteichoic acid synthase (LtaS) and the ß-lactam response sensor BlaR1 in the medium is dependent on SPase activity, suggesting that they are cleaved at noncanonical sites within the protein. In all, the data define the proteins whose stationary-phase secretion depends on SPase and also suggest that the arylomycins should be valuable chemical biology tools for the study of protein secretion in a wide variety of different bacteria.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Staphylococcus epidermidis/metabolism , Chromatography, Liquid , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mass Spectrometry , Membrane Proteins/antagonists & inhibitors , Molecular Structure , Oligopeptides/chemistry , Oligopeptides/pharmacology , Protein Sorting Signals/genetics
7.
Chem Biol ; 17(11): 1223-31, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21095572

ABSTRACT

Novel classes of broad-spectrum antibiotics are needed to treat multidrug-resistant pathogens. The arylomycin class of natural products inhibits a promising antimicrobial target, type I signal peptidase (SPase), but upon initial characterization appeared to lack whole-cell activity against most pathogens. Here, we show that Staphylococcus epidermidis, which is sensitive to the arylomycins, evolves resistance via mutations in SPase and that analogous mutations are responsible for the natural resistance of Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa. We identify diverse bacteria lacking these mutations and demonstrate that most are sensitive to the arylomycins. The results illustrate that the arylomycins have a broad-spectrum of activity and are viable candidates for development into therapeutics. The results also raise the possibility that naturally occurring resistance may have masked other natural product scaffolds that might be developed into therapeutics.


Subject(s)
Anti-Bacterial Agents/chemistry , Membrane Proteins/antagonists & inhibitors , Oligopeptides/chemistry , Amino Acid Substitution , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microbial Sensitivity Tests , Oligopeptides/pharmacology , Protein Structure, Tertiary , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Staphylococcus epidermidis/enzymology
8.
Microbiology (Reading) ; 156(Pt 10): 3031-3040, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20522495

ABSTRACT

The chlamydial protease/proteasome-like activity factor (CPAF) is secreted into the host cytosol to degrade various host factors that benefit chlamydial intracellular survival. Although the full-length CPAF is predicted to contain a putative signal peptide at its N terminus, the secretion pathway of CPAF is still unknown. Here, we have provided experimental evidence that the N-terminal sequence covering the M1-G31 region was cleaved from CPAF during chlamydial infection. The CPAF N-terminal sequence, when expressed in a phoA gene fusion construct, was able to direct the export of the mature PhoA protein across the inner membrane of wild-type Escherichia coli. However, E. coli mutants deficient in SecB failed to support the CPAF signal-peptide-directed secretion of PhoA. Since native PhoA secretion was known to be independent of SecB, this SecB dependence must be rendered by the CPAF leader peptide. Furthermore, lack of SecY function also blocked the CPAF signal-peptide-directed secretion of PhoA. Most importantly, CPAF secretion into the host cell cytosol during chlamydial infection was selectively inhibited by an inhibitor specifically targeting type I signal peptidase but not by a type III secretion-system-specific inhibitor. Together, these observations have demonstrated that the chlamydial virulence factor CPAF relies on Sec-dependent transport for crossing the chlamydial inner membrane, which has provided essential information for further delineating the pathways of CPAF action and understanding chlamydial pathogenic mechanisms.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/metabolism , Protein Sorting Signals , Virulence Factors/metabolism , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Chlamydia trachomatis/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Protein Transport , Virulence Factors/genetics
9.
J Am Chem Soc ; 129(51): 15830-8, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18052061

ABSTRACT

The growing threat of untreatable bacterial infections has refocused efforts to identify new antibiotics, especially those acting by novel mechanisms. While the inhibition of pathogen proteases has proven to be a successful strategy for drug development, such inhibitors are often limited by toxicity due to their promiscuous inhibition of homologous and mechanistically related human enzymes. Unlike many protease inhibitors, inhibitors of the essential type I bacterial signal peptidase (SPase) may be more specific and thus less toxic due to the enzyme's unique structure and catalytic mechanism. Recently, the arylomycins and related lipoglycopeptide natural products were isolated and shown to inhibit SPase. The core structure of the arylomycins and lipoglycopeptides consists of a biaryl-linked, N-methylated peptide macrocycle attached to a lipopeptide tail, and in the case of the lipoglycopeptides, a deoxymannose moiety. Herein, we report the first total synthesis of a member of this group of antibiotics, arylomycin A2. The synthesis relies on Suzuki-Miyaura-mediated biaryl coupling, which model studies suggested would be more efficient than a lactamization-based route. Biological studies demonstrate that these compounds are promising antibiotics, especially against Gram-positive pathogens, with activity against S. epidermidis that equals that of the currently prescribed antibiotics. Structural and biological studies suggest that both N-methylation and lipidation may contribute to antibiotic activity, whereas glycosylation appears to be generally less critical. Thus, these studies help identify the determinants of the biological activity of arylomycin A2 and should aid in the design of analogs to further explore and develop this novel class of antibiotic.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/pharmacology , Catalysis , Humans , Hydrogen Bonding , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...