Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Mol Microbiol ; 121(5): 1021-1038, 2024 May.
Article in English | MEDLINE | ID: mdl-38527904

ABSTRACT

Daptomycin is a last-line antibiotic commonly used to treat vancomycin-resistant Enterococci, but resistance evolves rapidly and further restricts already limited treatment options. While genetic determinants associated with clinical daptomycin resistance (DAPR) have been described, information on factors affecting the speed of DAPR acquisition is limited. The multiple peptide resistance factor (MprF), a phosphatidylglycerol-modifying enzyme involved in cationic antimicrobial resistance, is linked to DAPR in pathogens such as methicillin-resistant Staphylococcus aureus. Since Enterococcus faecalis encodes two paralogs of mprF and clinical DAPR mutations do not map to mprF, we hypothesized that functional redundancy between the paralogs prevents mprF-mediated resistance and masks other evolutionary pathways to DAPR. Here, we performed in vitro evolution to DAPR in mprF mutant background. We discovered that the absence of mprF results in slowed DAPR evolution and is associated with inactivating mutations in ftsH, resulting in the depletion of the chaperone repressor HrcA. We also report that ftsH is essential in the parental, but not in the ΔmprF, strain where FtsH depletion results in growth impairment in the parental strain, a phenotype associated with reduced extracellular acidification and reduced ability for metabolic reduction. This presents FtsH and HrcA as enticing targets for developing anti-resistance strategies.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Daptomycin , Enterococcus faecalis , Microbial Sensitivity Tests , Enterococcus faecalis/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/metabolism , Enterococcus faecalis/enzymology , Daptomycin/pharmacology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology , Mutation , Drug Resistance, Bacterial/genetics , Peptide Hydrolases/metabolism , Peptide Hydrolases/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism
2.
Sci Transl Med ; 16(735): eadi7558, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38381846

ABSTRACT

Infections caused by nontuberculous mycobacteria have increased more than 50% in the past two decades and more than doubled in the elderly population. Mycobacterium abscessus (Mab), one of the most prevalent of these rapidly growing species, is intrinsically resistant to numerous antibiotics. Current standard-of-care treatments are not satisfactory, with high failure rate and notable adverse effects. We report here a potent anti-Mab compound from the flexible molecular framework afforded by conjugated oligoelectrolytes (COEs). A screen of structurally diverse, noncytotoxic COEs identified a lead compound, COE-PNH2, which was bactericidal against replicating, nonreplicating persisters and intracellular Mab.COE-PNH2 had low propensity for resistance development, with a frequency of resistance below 1.25 × 10-9 and showed no detectable resistance upon serial passaging. Mechanism of action studies were in line with COE-PNH2 affecting the physical and functional integrity of the bacterial envelope and disrupting the mycomembrane and associated essential bioenergetic pathways. Moreover, COE-PNH2 was well-tolerated and efficacious in a mouse model of Mab lung infection. This study highlights desirable in vitro and in vivo potency and safety index of this COE structure, which represents a promising anti-mycobacterial to tackle an unmet medical need.


Subject(s)
Mycobacterium abscessus , Mycobacterium , Humans , Aged , Animals , Mice , Disease Models, Animal , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests
3.
J Biol Chem ; 300(2): 105618, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38176652

ABSTRACT

The F1FO-ATP synthase engine is essential for viability and growth of nontuberculous mycobacteria (NTM) by providing the biological energy ATP and keeping ATP homeostasis under hypoxic stress conditions. Here, we report the discovery of the diarylquinoline TBAJ-5307 as a broad spectrum anti-NTM inhibitor, targeting the FO domain of the engine and preventing rotation and proton translocation. TBAJ-5307 is active at low nanomolar concentrations against fast- and slow-growing NTM as well as clinical isolates by depleting intrabacterial ATP. As demonstrated for the fast grower Mycobacterium abscessus, the compound is potent in vitro and in vivo, without inducing toxicity. Combining TBAJ-5307 with anti-NTM antibiotics or the oral tebipenem-avibactam pair showed attractive potentiation. Furthermore, the TBAJ-5307-tebipenem-avibactam cocktail kills the pathogen, suggesting a novel oral combination for the treatment of NTM lung infections.


Subject(s)
Anti-Bacterial Agents , Diarylquinolines , Enzyme Inhibitors , Mycobacterium Infections, Nontuberculous , Nontuberculous Mycobacteria , Humans , Adenosine Triphosphate , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Azabicyclo Compounds , Carbapenems , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Diarylquinolines/pharmacology
4.
Microbiol Spectr ; 11(6): e0228223, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37982630

ABSTRACT

IMPORTANCE: New drugs are needed to combat multidrug-resistant tuberculosis. The electron transport chain (ETC) maintains the electrochemical potential across the cytoplasmic membrane and allows the production of ATP, the energy currency of any living cell. The mycobacterial engine F-ATP synthase catalyzes the formation of ATP and has come into focus as an attractive and rich drug target. Recent deep insights into these mycobacterial F1FO-ATP synthase elements opened the door for a renaissance of structure-based target identification and inhibitor design. In this study, we present the GaMF1.39 antimycobacterial compound, targeting the rotary subunit γ of the biological engine. The compound is bactericidal, inhibits infection ex vivo, and displays enhanced anti-tuberculosis activity in combination with ETC inhibitors, which promises new strategies to shorten tuberculosis chemotherapy.


Subject(s)
Clofazimine , Mycobacterium tuberculosis , Clofazimine/pharmacology , Clofazimine/therapeutic use , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Adenosine Triphosphate
5.
Expert Opin Drug Discov ; 18(8): 917-927, 2023.
Article in English | MEDLINE | ID: mdl-37332221

ABSTRACT

INTRODUCTION: Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is the most devastating bacterial disease. Multidrug-resistant Mtb strains are spreading worldwide, underscoring the need for new anti-TB targets and inhibitors. The respiratory chain complexes, including the cytochrome bd oxidase (cyt-bd), have been identified as an attractive target for drug development. Recent novel structural and mechanistic insight as well as inhibitors of Mtb's cyt-bd brought this enzyme into the focus. AREAS COVERED: In this review, the authors describe conditions that stimulate the biogenesis of Mtb cyt-bd, its structural-, mechanistic-, and substrate-binding traits. They discuss the present Mtb cyt-bd inhibitors, novel targets within the enzyme and structure activity relationship features that are required for mycobacterial cyt-bd inhibition and augment their understanding on improving the potency of cyt-bd inhibitors. EXPERT OPINION: A deeper structure-mechanistic understanding of Mtb's cyt-bd is a prerequisite for in silico efforts to: (i) identify pathogen specific targets for the design of novel nontoxic hit molecules, forming the platform for the development of new leads, (ii) design mechanism of action studies, (iii) perform medicinal chemistry of existing inhibitors to improve their potency and pharmacokinetic/-dynamic properties. Phase studies with such optimized cyt-bd inhibitors in combination with anti-TB compounds targeting the oxidative phosphorylation pathway is recommended.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Electron Transport Complex IV/metabolism , Tuberculosis/drug therapy , Tuberculosis/microbiology , Cytochromes
6.
Antimicrob Agents Chemother ; 67(6): e0153122, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37158740

ABSTRACT

The mycobacterial cytochrome bcc:aa3 complex deserves the name "supercomplex" since it combines three cytochrome oxidases-cytochrome bc, cytochrome c, and cytochrome aa3-into one supramolecular machine and performs electron transfer for the reduction of oxygen to water and proton transport to generate the proton motive force for ATP synthesis. Thus, the bcc:aa3 complex represents a valid drug target for Mycobacterium tuberculosis infections. The production and purification of an entire M. tuberculosis cytochrome bcc:aa3 are fundamental for biochemical and structural characterization of this supercomplex, paving the way for new inhibitor targets and molecules. Here, we produced and purified the entire and active M. tuberculosis cyt-bcc:aa3 oxidase, as demonstrated by the different heme spectra and an oxygen consumption assay. The resolved M. tuberculosis cyt-bcc:aa3 cryo-electron microscopy structure reveals a dimer with its functional domains involved in electron, proton, oxygen transfer, and oxygen reduction. The structure shows the two cytochrome cIcII head domains of the dimer, the counterpart of the soluble mitochondrial cytochrome c, in a so-called "closed state," in which electrons are translocated from the bcc to the aa3 domain. The structural and mechanistic insights provided the basis for a virtual screening campaign that identified a potent M. tuberculosis cyt-bcc:aa3 inhibitor, cytMycc1. cytMycc1 targets the mycobacterium-specific α3-helix of cytochrome cI and interferes with oxygen consumption by interrupting electron translocation via the cIcII head. The successful identification of a new cyt-bcc:aa3 inhibitor demonstrates the potential of a structure-mechanism-based approach for novel compound development.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Cryoelectron Microscopy , Cytochromes c , Protons , Oxygen
7.
JAC Antimicrob Resist ; 5(3): dlad052, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37168836

ABSTRACT

Background: Mycobacterium abscessus is a non-tuberculous mycobacterium (NTM) that causes chronic pulmonary infections. Because of its extensive innate resistance to numerous antibiotics, treatment options are limited, often resulting in poor clinical outcomes. Current treatment regimens usually involve a combination of antibiotics, with clarithromycin being the cornerstone of NTM treatments. Objectives: To identify drug candidates that exhibit synergistic activity with clarithromycin against M. abscessus. Methods: We performed cell-based phenotypic screening of a compound library against M. abscessus induced to become resistant to clarithromycin. Furthermore, we evaluated the toxicity and efficacy of the top compound in a zebrafish embryo infection model. Results: The screen revealed rifaximin as a clarithromycin potentiator. The combination of rifaximin and clarithromycin was synergistic and bactericidal in vitro and potent in the zebrafish model. Conclusions: The data indicate that the rifaximin/clarithromycin combination is promising to effectively treat pulmonary NTM infections.

8.
Curr Opin Microbiol ; 74: 102327, 2023 08.
Article in English | MEDLINE | ID: mdl-37235914

ABSTRACT

The approval of the first-in-class antibacterial bedaquiline for tuberculosis marks a breakthrough in antituberculosis drug development. The drug inhibits mycobacterial respiration and represents the validation of a wholly different metabolic process as a druggable target space. In this review, we discuss the advances in the development of mycobacterial respiratory inhibitors, as well as the potential of applying this strategy to other pathogens. The non-fermentative nature of mycobacteria explains their vulnerability to respiration inhibition, and we caution that this strategy may not be equally effective in other organisms. Conversely, we also showcase fundamental studies that reveal ancillary functions of the respiratory pathway, which are crucial to some pathogens' virulence, drug susceptibility and fitness, introducing another perspective of targeting bacterial respiration as an antibiotic strategy.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Tuberculosis/drug therapy , Respiration , Mycobacterium tuberculosis/genetics
9.
Chem Sci ; 14(16): 4434, 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37123184

ABSTRACT

[This corrects the article DOI: 10.1039/D1SC05835E.].

10.
Cell Rep ; 42(5): 112444, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37115669

ABSTRACT

The bioenergetic mechanisms by which Mycobacterium tuberculosis survives hypoxia are poorly understood. Current models assume that the bacterium shifts to an alternate electron acceptor or fermentation to maintain membrane potential and ATP synthesis. Counterintuitively, we find here that oxygen itself is the principal terminal electron acceptor during hypoxic dormancy. M. tuberculosis can metabolize oxygen efficiently at least two orders of magnitude below the concentration predicted to occur in hypoxic lung granulomas. Despite a difference in apparent affinity for oxygen, both the cytochrome bcc:aa3 and cytochrome bd oxidase respiratory branches are required for hypoxic respiration. Simultaneous inhibition of both oxidases blocks oxygen consumption, reduces ATP levels, and kills M. tuberculosis under hypoxia. The capacity of mycobacteria to scavenge trace levels of oxygen, coupled with the absence of complex regulatory mechanisms to achieve hierarchal control of the terminal oxidases, may be a key determinant of long-term M. tuberculosis survival in hypoxic lung granulomas.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Mycobacterium tuberculosis/metabolism , Oxygen/metabolism , Electron Transport Complex IV/metabolism , Oxidoreductases/metabolism , Homeostasis , Tuberculosis/microbiology , Hypoxia , Adenosine Triphosphate/metabolism , Cytochromes/metabolism
11.
JACS Au ; 3(2): 276-292, 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36873689

ABSTRACT

The antimicrobial resistance crisis is a global health issue requiring discovery and development of novel therapeutics. However, conventional screening of natural products or synthetic chemical libraries is uncertain. Combination therapy using approved antibiotics with inhibitors targeting innate resistance mechanisms provides an alternative strategy to develop potent therapeutics. This review discusses the chemical structures of effective ß-lactamase inhibitors, outer membrane permeabilizers, and efflux pump inhibitors that act as adjuvant molecules of classical antibiotics. Rational design of the chemical structures of adjuvants will provide methods to impart or restore efficacy to classical antibiotics for inherently antibiotic-resistant bacteria. As many bacteria have multiple resistance pathways, adjuvant molecules simultaneously targeting multiple pathways are promising approaches to combat multidrug-resistant bacterial infections.

12.
Biomaterials ; 294: 122004, 2023 03.
Article in English | MEDLINE | ID: mdl-36669302

ABSTRACT

New antimicrobials are urgently needed to combat Gram-negative bacteria, particularly multi-drug resistant (MDR) and phenotypically resistant biofilm species. At present, only sequence-defined alpha-peptides (e.g. polymyxin B) can selectively target Gram-negative bacterial lipopolysaccharides. We show that a copolymer, without a defined sequence, shows good potency against MDR Gram-negative bacteria including its biofilm form. The tapered blocky co-beta-peptide with controlled N-terminal hydrophobicity (#4) has strong interaction with the Gram-negative bacterial lipopolysaccharides via its backbone through electrostatic and hydrogen bonding interactions but not the Gram-positive bacterial and mammalian cell membranes so that this copolymer is non-toxic to these two latter cell types. The new #4 co-beta-peptide selectively kills Gram-negative bacteria with low cytotoxicity both in vitro and in a mouse biofilm wound infection model. This strategy provides a new concept for the design of Gram-negative selective antimicrobial peptidomimetics against MDR and biofilm species.


Subject(s)
Anti-Infective Agents , Peptides , Animals , Mice , Gram-Negative Bacteria/metabolism , Lipopolysaccharides , Biofilms , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Mammals/metabolism
13.
Cell Chem Biol ; 29(8): 1317-1324.e5, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35901793

ABSTRACT

New therapeutic concepts are critically needed for carbapenem-resistant Pseudomonas aeruginosa, an opportunistic pathogen particularly recalcitrant to antibiotics. The screening of around 230,000 small molecules yielded a very low hit rate of 0.002% after triaging for known antibiotics. The only novel hit that stood out was the antimetabolite oxythiamine. Oxythiamine is a known transketolase inhibitor in eukaryotic cells, but its antibacterial potency has not been reported. Metabolic and transcriptomic analyses indicated that oxythiamine is intracellularly converted to oxythiamine pyrophosphate and subsequently inhibits several vitamin-B1-dependent enzymes, sensitizing the bacteria to several antibiotic and non-antibiotic drugs such as tetracyclines, 5-fluorouracil, and auranofin. The positive interaction between 5-fluorouracil and oxythiamine was confirmed in a murine ocular infection model, indicating relevance during infection. Together, this study revealed a system-level significance of thiamine metabolism perturbation that sensitizes P. aeruginosa to multiple small molecules, a property that could inform on the development of a rational drug combination.


Subject(s)
Oxythiamine , Thiamine Pyrophosphate , Animals , Anti-Bacterial Agents/pharmacology , Fluorouracil , Mice , Oxythiamine/metabolism , Oxythiamine/pharmacology , Pseudomonas aeruginosa/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Thiamine/metabolism , Thiamine/pharmacology , Thiamine Pyrophosphate/analysis , Thiamine Pyrophosphate/metabolism
14.
Chem Sci ; 13(2): 345-364, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35126968

ABSTRACT

The growing prevalence of antimicrobial drug resistance in pathogenic bacteria is a critical threat to global health. Conventional antibiotics still play a crucial role in treating bacterial infections, but the emergence and spread of antibiotic-resistant micro-organisms are rapidly eroding their usefulness. Cationic polymers, which target bacterial membranes, are thought to be the last frontier in antibacterial development. This class of molecules possesses several advantages including a low propensity for emergence of resistance and rapid bactericidal effect. This review surveys the structure-activity of advanced antimicrobial cationic polymers, including poly(α-amino acids), ß-peptides, polycarbonates, star polymers and main-chain cationic polymers, with low toxicity and high selectivity to potentially become useful for real applications. Their uses as potentiating adjuvants to overcome bacterial membrane-related resistance mechanisms and as antibiofilm agents are also covered. The review is intended to provide valuable information for design and development of cationic polymers as antimicrobial and antibiofilm agents for translational applications.

15.
Emerg Infect Dis ; 28(3): 749-751, 2022 03.
Article in English | MEDLINE | ID: mdl-35202539

ABSTRACT

The treatment of leprosy is long and complex, benefiting from the development of sterilizing, rapidly-acting drugs. Reductive evolution made Mycobacterium leprae exquisitely sensitive to Telacebec, a phase 2 drug candidate for tuberculosis. The unprecedented potency of Telacebec against M. leprae warrants further validation in clinical trials.


Subject(s)
Mycobacterium leprae , Pyridines , Imidazoles , Piperidines
16.
Expert Opin Investig Drugs ; 31(2): 139-144, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35034512

ABSTRACT

INTRODUCTION: Tuberculosis is an infectious disease that affected more than 50 million people and killed 6.7 million patients in the past 5 years alone. Additionally, rising incidence of treatment resistance threatens the global effort to eradicate this disease. With limited options available, additional novel antibiotics are needed for the treatment of multidrug-resistant tuberculosis (MDR-TB). Telacebec is a first-in-class antibiotic that targets the pathogen's energy metabolism. AREAS COVERED: This paper provides an overview of the recent progress in the development and testing of telacebec. We discuss published clinical data and examine the design and setup of its clinical trials. We also offer insights on the therapeutic potential of telacebec and aspects of which should be evaluated in the future. EXPERT OPINION: The first phase 2a trial showed a correlation between dosage and bacterial load in patient sputum, which should be confirmed using a direct measurement method such as colony-forming unit counting. Its clinical efficacy, favorable pharmacokinetic properties, low arrhythmogenic risk, and activity against MDR-TB strains make telacebec a suitable candidate for further development. Future clinical testing in combination with approved second-line drugs will reveal its full potential against MDR-TB. Considering recent preclinical studies, we also recommend initiating clinical trials for Buruli ulcer and leprosy.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis, Multidrug-Resistant , Tuberculosis , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Humans , Imidazoles , Piperidines/therapeutic use , Pyridines/therapeutic use , Tuberculosis/drug therapy , Tuberculosis, Multidrug-Resistant/drug therapy
17.
Methods Mol Biol ; 2387: 209-217, 2022.
Article in English | MEDLINE | ID: mdl-34643915

ABSTRACT

Generation and characterization of drug resistant mutants is a powerful tool in antimicrobial drug discovery for identification of the molecular target of an investigational drug candidate. The method is relatively simple to be conducted in a classical microbiology laboratory. Its value has been augmented by the employment of next generation sequencing techniques to characterize single-nucleotide polymorphisms associated with drug resistance. Determination of the frequency of emergence of resistance to drug candidates also provides insights into their usefulness for clinical application. In addition to the generation of drug resistant mutants, we describe a direct method to determine the minimum inhibitory concentration of a drug candidate against Mycobacterium ulcerans.


Subject(s)
Mycobacterium ulcerans , Anti-Bacterial Agents , Buruli Ulcer , Humans , Microbial Sensitivity Tests , Mycobacterium ulcerans/genetics , Pharmaceutical Preparations
18.
Methods Mol Biol ; 2387: 219-230, 2022.
Article in English | MEDLINE | ID: mdl-34643916

ABSTRACT

The oxidative phosphorylation (OxPhos) pathway has emerged as an attractive pathway for the development of anti-mycobacterial drugs. The OxPhos pathway is essential for ATP resynthesis and maintenance of the electrochemical transmembrane gradient. The bioenergetic parameters of the pathway such as oxygen consumption rate and ATP levels are quantifiable using current technology. Measuring these parameters are useful tools to gauge rapidly the impact of drug candidates on their capacity to inhibit the OxPhos pathway in Mycobacterium ulcerans.


Subject(s)
Mycobacterium ulcerans , Adenosine Triphosphate , Buruli Ulcer , Energy Metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...