Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Biol Drug Des ; 103(6): e14569, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38877369

ABSTRACT

Staphylococcus aureus has the ability to invade cortical bone osteocyte lacuno-canalicular networks (OLCNs) and cause osteomyelitis. It was recently established that the cell wall transpeptidase, penicillin-binding protein 4 (PBP4), is crucial for this function, with pbp4 deletion strains unable to invade OLCNs and cause bone pathogenesis in a murine model of S. aureus osteomyelitis. Moreover, PBP4 has recently been found to modulate S. aureus resistance to ß-lactam antibiotics. As such, small molecule inhibitors of S. aureus PBP4 may represent dual functional antimicrobial agents that limit osteomyelitis and/or reverse antibiotic resistance. A high throughput screen recently revealed that the phenyl-urea 1 targets PBP4. Herein, we describe a structure-activity relationship (SAR) study on 1. Leveraging in silico docking and modeling, a set of analogs was synthesized and assessed for PBP4 inhibitory activities. Results revealed a preliminary SAR and identified lead compounds with enhanced binding to PBP4, more potent antibiotic resistance reversal, and diminished PBP4 cell wall transpeptidase activity in comparison to 1.


Subject(s)
Anti-Bacterial Agents , Molecular Docking Simulation , Penicillin-Binding Proteins , Staphylococcus aureus , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Structure-Activity Relationship , Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Urea/chemistry , Urea/pharmacology , Urea/analogs & derivatives , Animals , Mice , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors
2.
Cell Rep ; 42(6): 112540, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37227819

ABSTRACT

Pseudomonas aeruginosa and Staphylococcus aureus are among the most frequently isolated bacterial species from polymicrobial infections of patients with cystic fibrosis and chronic wounds. We apply mass spectrometry guided interaction studies to determine how chemical interaction shapes the fitness and community structure during co-infection of these two pathogens. We demonstrate that S. aureus is equipped with an elegant mechanism to inactivate pyochelin via the yet uncharacterized methyltransferase Spm (staphylococcal pyochelin methyltransferase). Methylation of pyochelin abolishes the siderophore activity of pyochelin and significantly lowers pyochelin-mediated intracellular reactive oxygen species (ROS) production in S. aureus. In a murine wound co-infection model, an S. aureus mutant unable to methylate pyochelin shows significantly lower fitness compared with its parental strain. Thus, Spm-mediated pyochelin methylation is a mechanism to increase S. aureus survival during in vivo competition with P. aeruginosa.


Subject(s)
Coinfection , Staphylococcal Infections , Humans , Mice , Animals , Staphylococcus aureus/physiology , Pseudomonas aeruginosa/metabolism , Coinfection/microbiology , Staphylococcal Infections/microbiology
3.
Int J Mol Sci ; 23(6)2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35328372

ABSTRACT

Biofilm growth is thought to be a significant obstacle to the successful treatment of Mycobacterium abscessus infections. A search for agents capable of inhibiting M. abscessus biofilms led to our interest in 2-aminoimidazoles and related scaffolds, which have proven to display antibiofilm properties against a number of Gram-negative and Gram-positive bacteria, including Mycobacterium tuberculosis and Mycobacterium smegmatis. The screening of a library of 30 compounds led to the identification of a compound, AB-2-29, which inhibits the formation of M. abscessus biofilms with an IC50 (the concentration required to inhibit 50% of biofilm formation) in the range of 12.5 to 25 µM. Interestingly, AB-2-29 appears to chelate zinc, and its antibiofilm activity is potentiated by the addition of zinc to the culture medium. Preliminary mechanistic studies indicate that AB-2-29 acts through a distinct mechanism from those reported to date for 2-aminoimidazole compounds.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Anti-Bacterial Agents/pharmacology , Biofilms , Humans , Imidazoles/pharmacology , Microbial Sensitivity Tests , Zinc/pharmacology
4.
Angew Chem Int Ed Engl ; 61(17): e202117458, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35167718

ABSTRACT

Over the past decades, antibiotic resistance has grown to a point where orthogonal approaches to combating infections caused by resistant bacteria are needed. One such approach is the development of non-microbicidal small molecules that potentiate the activity of conventional antibiotics, termed adjuvants. The diterpene natural product 12(S),16ϵ-dihydroxycleroda-3,13-dien-15,16-olide, which we refer to as (-)-LZ-2112, is known to synergize with oxacillin against methicillin-resistant Staphylococcus aureus (MRSA). To explore this activity, (-)-LZ-2112 was synthesized and the structure confirmed through X-ray analysis. Preliminary structure-activity relationship studies following the synthesis of several analogs identified key structural elements responsible for activity and indicate that scaffold simplification is possible. A preliminary mode of action study suggests mecA plays a role in the adjuvant activity of (-)-LZ-2112.


Subject(s)
Diterpenes, Clerodane , Methicillin-Resistant Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Diterpenes, Clerodane/pharmacology , Microbial Sensitivity Tests , Oxacillin/pharmacology , beta-Lactams/pharmacology
5.
ChemMedChem ; 15(17): 1672-1679, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32662926

ABSTRACT

Drug-resistant bacteria are rapidly becoming a significant problem across the globe. One element that factors into this crisis is the role played by bacterial biofilms in the recalcitrance of some infections to the effects of conventional antibiotics. Bacteria within a biofilm are highly tolerant of both antibiotic treatment and host immune responses. Biofilms are implicated in many chronic infections, including tuberculosis, in which they can act as bacterial reservoirs, requiring an arduous antibiotic regimen to eradicate the infection. A separate, compounding problem is that antibiotics once seen as last-resort drugs, such as the polymyxin colistin, are now seeing more frequent usage as resistance to front-line drugs in Gram-negative bacteria becomes more prevalent. The increased use of such antibiotics inevitably leads to an increased frequency of resistance. Drugs that inhibit biofilms and/or act as adjuvants to overcome resistance to existing antibiotics will potentially be an important component of future approaches to antibacterial treatment. We have previously demonstrated that analogues of the meridianin natural product family possess adjuvant and antibiofilm activities. In this study, we explore structural variation of the lead molecule from previous studies, and identify compounds showing both improved biofilm inhibition potency and synergy with colistin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biological Products/pharmacology , Colistin/pharmacology , Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Colistin/chemistry , Dose-Response Relationship, Drug , Drug Resistance, Multiple, Bacterial/drug effects , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium smegmatis/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL