Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Mol Biosci ; 9: 880432, 2022.
Article in English | MEDLINE | ID: mdl-35712348

ABSTRACT

Anti-microbial resistance is a rising global healthcare concern that needs urgent attention as growing number of infections become difficult to treat with the currently available antibiotics. This is particularly true for mycobacterial infections like tuberculosis and leprosy and those with emerging opportunistic pathogens such as Mycobacterium abscessus, where multi-drug resistance leads to increased healthcare cost and mortality. M. abscessus is a highly drug-resistant non-tuberculous mycobacterium which causes life-threatening infections in people with chronic lung conditions such as cystic fibrosis. In this study, we explore M. abscessus phosphopantetheine adenylyl transferase (PPAT), an enzyme involved in the biosynthesis of Coenzyme A, as a target for the development of new antibiotics. We provide structural insights into substrate and feedback inhibitor binding modes of M. abscessus PPAT, thereby setting the basis for further chemical exploration of the enzyme. We then utilize a multi-dimensional fragment screening approach involving biophysical and structural analysis, followed by evaluation of compounds from a previous fragment-based drug discovery campaign against M. tuberculosis PPAT ortholog. This allowed the identification of an early-stage lead molecule exhibiting low micro molar affinity against M. abscessus PPAT (Kd 3.2 ± 0.8 µM) and potential new ways to design inhibitors against this enzyme. The resulting crystal structures reveal striking conformational changes and closure of solvent channel of M. abscessus PPAT hexamer providing novel strategies of inhibition. The study thus validates the ligandability of M. abscessus PPAT as an antibiotic target and identifies crucial starting points for structure-guided drug discovery against this bacterium.

2.
ACS Infect Dis ; 8(2): 296-309, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35037462

ABSTRACT

Mycobacterium abscessus (Mab) has emerged as a challenging threat to individuals with cystic fibrosis. Infections caused by this pathogen are often impossible to treat due to the intrinsic antibiotic resistance leading to lung malfunction and eventually death. Therefore, there is an urgent need to develop new drugs against novel targets in Mab to overcome drug resistance and subsequent treatment failure. In this study, SAICAR synthetase (PurC) from Mab was identified as a promising target for novel antibiotics. An in-house fragment library screen and a high-throughput X-ray crystallographic screen of diverse fragment libraries were explored to provide crucial starting points for fragment elaboration. A series of compounds developed from fragment growing and merging strategies, guided by crystallographic information and careful hit-to-lead optimization, have achieved potent nanomolar binding affinity against the enzyme. Some compounds also show a promising inhibitory effect against Mab and Mtb. This work utilizes a fragment-based design and demonstrates for the first time the potential to develop inhibitors against PurC from Mab.


Subject(s)
Mycobacterium abscessus , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , Humans , Peptide Synthases
3.
J Med Chem ; 65(3): 2149-2173, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35080396

ABSTRACT

Pseudomonas aeruginosa is of major concern for cystic fibrosis patients where this infection can be fatal. With the emergence of drug-resistant strains, there is an urgent need to develop novel antibiotics against P. aeruginosa. MurB is a promising target for novel antibiotic development as it is involved in the cell wall biosynthesis. MurB has been shown to be essential in P. aeruginosa, and importantly, no MurB homologue exists in eukaryotic cells. A fragment-based drug discovery approach was used to target Pa MurB. This led to the identification of a number of fragments, which were shown to bind to MurB. One fragment, a phenylpyrazole scaffold, was shown by ITC to bind with an affinity of Kd = 2.88 mM (LE 0.23). Using a structure guided approach, different substitutions were synthesized and the initial fragment was optimized to obtain a small molecule with Kd = 3.57 µM (LE 0.35).


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Pseudomonas aeruginosa/enzymology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cystic Fibrosis/complications , Cystic Fibrosis/mortality , Cystic Fibrosis/pathology , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Conformation , Molecular Docking Simulation , Oxidoreductases/metabolism , Pseudomonas Infections/complications , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use
4.
Front Microbiol ; 12: 706207, 2021.
Article in English | MEDLINE | ID: mdl-34335541

ABSTRACT

Chronic pulmonary infections caused by non-tuberculous mycobacteria of the Mycobacterium abscessus complex (MABSC) are emerging as a global health problem and pose a threat to susceptible individuals with structural lung disease such as cystic fibrosis. The molecular mechanisms underlying the pathogenicity and intrinsic resistance of MABSC to antibiotics remain largely unknown. The involvement of Msp-type porins in the virulence and biocide resistance of some rapidly growing non-tuberculous mycobacteria and the finding of deletions and rearrangements in the porin genes of serially collected MABSC isolates from cystic fibrosis patients prompted us to investigate the contribution of these major surface proteins to MABSC infection. Inactivation by allelic replacement of the each of the two Msp-type porin genes of M. abscessus subsp. massiliense CIP108297, mmpA and mmpB, led to a marked increase in the virulence and pathogenicity of both mutants in murine macrophages and infected mice. Neither of the mutants were found to be significantly more resistant to antibiotics. These results suggest that adaptation to the host environment rather than antibiotic pressure is the key driver of the emergence of porin mutants during infection.

5.
Science ; 372(6541)2021 04 30.
Article in English | MEDLINE | ID: mdl-33926925

ABSTRACT

Although almost all mycobacterial species are saprophytic environmental organisms, a few, such as Mycobacterium tuberculosis, have evolved to cause transmissible human infection. By analyzing the recent emergence and spread of the environmental organism M. abscessus through the global cystic fibrosis population, we have defined key, generalizable steps involved in the pathogenic evolution of mycobacteria. We show that epigenetic modifiers, acquired through horizontal gene transfer, cause saltational increases in the pathogenic potential of specific environmental clones. Allopatric parallel evolution during chronic lung infection then promotes rapid increases in virulence through mutations in a discrete gene network; these mutations enhance growth within macrophages but impair fomite survival. As a consequence, we observe constrained pathogenic evolution while person-to-person transmission remains indirect, but postulate accelerated pathogenic adaptation once direct transmission is possible, as observed for M. tuberculosis Our findings indicate how key interventions, such as early treatment and cross-infection control, might restrict the spread of existing mycobacterial pathogens and prevent new, emergent ones.


Subject(s)
Communicable Diseases, Emerging/microbiology , Evolution, Molecular , Genetic Fitness , Lung/microbiology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium abscessus/genetics , Mycobacterium abscessus/pathogenicity , Pneumonia, Bacterial/microbiology , Communicable Diseases, Emerging/transmission , Datasets as Topic , Epigenesis, Genetic , Gene Transfer, Horizontal , Genome, Bacterial , Humans , Mutation , Mycobacterium Infections, Nontuberculous/transmission , Pneumonia, Bacterial/transmission , Virulence/genetics
6.
ACS Infect Dis ; 6(8): 2143-2154, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32551551

ABSTRACT

Understanding the physiological processes underlying the ability of Mycobacterium abscessus to become a chronic pathogen of the cystic fibrosis (CF) lung is important to the development of prophylactic and therapeutic strategies to better control and treat pulmonary infections caused by these bacteria. Gene expression profiling of a diversity of M. abscessus complex isolates points to amino acids being significant sources of carbon and energy for M. abscessus in both CF sputum and synthetic CF medium and to the bacterium undergoing an important metabolic reprogramming in order to adapt to this particular nutritional environment. Cell envelope analyses conducted on the same representative isolates further revealed unexpected structural alterations in major cell surface glycolipids known as the glycopeptidolipids (GPLs). Besides showing an increase in triglycosylated forms of these lipids, CF sputum- and synthetic CF medium-grown isolates presented as yet unknown forms of GPLs representing as much as 10% to 20% of the total GPL content of the cells, in which the classical amino alcohol located at the carboxy terminal of the peptide, alaninol, is replaced with the branched-chain amino alcohol leucinol. Importantly, both these lipid changes were exacerbated by the presence of mucin in the culture medium. Collectively, our results reveal potential new drug targets against M. abscessus in the CF airway and point to mucin as an important host signal modulating the cell surface composition of this pathogen.


Subject(s)
Cystic Fibrosis , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Glycolipids , Humans , Mycobacterium abscessus/genetics , Sputum
7.
J Med Chem ; 62(15): 7210-7232, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31282680

ABSTRACT

Mycobacterium abscessus (Mab) is a rapidly growing species of multidrug-resistant nontuberculous mycobacteria that has emerged as a growing threat to individuals with cystic fibrosis and other pre-existing chronic lung diseases. Mab pulmonary infections are difficult, or sometimes impossible, to treat and result in accelerated lung function decline and premature death. There is therefore an urgent need to develop novel antibiotics with improved efficacy. tRNA (m1G37) methyltransferase (TrmD) is a promising target for novel antibiotics. It is essential in Mab and other mycobacteria, improving reading frame maintenance on the ribosome to prevent frameshift errors. In this work, a fragment-based approach was employed with the merging of two fragments bound to the active site, followed by structure-guided elaboration to design potent nanomolar inhibitors against Mab TrmD. Several of these compounds exhibit promising activity against mycobacterial species, including Mycobacterium tuberculosis and Mycobacterium leprae in addition to Mab, supporting the use of TrmD as a target for the development of antimycobacterial compounds.


Subject(s)
Anti-Bacterial Agents/chemistry , Drug Development/methods , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/enzymology , tRNA Methyltransferases/antagonists & inhibitors , tRNA Methyltransferases/metabolism , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray/methods , Humans , Protein Structure, Secondary
8.
Nat Commun ; 8: 14642, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262681

ABSTRACT

Mycobacterium tuberculosis remains a global threat to human health, yet the molecular mechanisms regulating immunity remain poorly understood. Cytokines can promote or inhibit mycobacterial survival inside macrophages and the underlying mechanisms represent potential targets for host-directed therapies. Here we show that cytokine-STAT signalling promotes mycobacterial survival within macrophages by deregulating lipid droplets via ATG2 repression. In Drosophila infected with Mycobacterium marinum, mycobacterium-induced STAT activity triggered by unpaired-family cytokines reduces Atg2 expression, permitting deregulation of lipid droplets. Increased Atg2 expression or reduced macrophage triglyceride biosynthesis, normalizes lipid deposition in infected phagocytes and reduces numbers of viable intracellular mycobacteria. In human macrophages, addition of IL-6 promotes mycobacterial survival and BCG-induced lipid accumulation by a similar, but probably not identical, mechanism. Our results reveal Atg2 regulation as a mechanism by which cytokines can control lipid droplet homeostasis and consequently resistance to mycobacterial infection in Drosophila.


Subject(s)
Autophagy-Related Proteins/immunology , Drosophila Proteins/immunology , Interleukin-6/metabolism , Mycobacterium Infections/immunology , STAT Transcription Factors/immunology , Vesicular Transport Proteins/immunology , Animals , Autophagy-Related Proteins/metabolism , Cell Line , Disease Models, Animal , Disease Resistance/immunology , Drosophila , Drosophila Proteins/metabolism , Hemocytes , Humans , Interleukin-6/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mycobacterium Infections/microbiology , Mycobacterium bovis/immunology , Mycobacterium bovis/pathogenicity , Mycobacterium marinum/immunology , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Primary Cell Culture , STAT Transcription Factors/metabolism , Signal Transduction/immunology , Triglycerides/immunology , Triglycerides/metabolism , Vesicular Transport Proteins/metabolism , Virulence
9.
Science ; 354(6313): 751-757, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27846606

ABSTRACT

Lung infections with Mycobacterium abscessus, a species of multidrug-resistant nontuberculous mycobacteria, are emerging as an important global threat to individuals with cystic fibrosis (CF), in whom M. abscessus accelerates inflammatory lung damage, leading to increased morbidity and mortality. Previously, M. abscessus was thought to be independently acquired by susceptible individuals from the environment. However, using whole-genome analysis of a global collection of clinical isolates, we show that the majority of M. abscessus infections are acquired through transmission, potentially via fomites and aerosols, of recently emerged dominant circulating clones that have spread globally. We demonstrate that these clones are associated with worse clinical outcomes, show increased virulence in cell-based and mouse infection models, and thus represent an urgent international infection challenge.


Subject(s)
Communicable Diseases, Emerging/microbiology , Cystic Fibrosis/microbiology , Drug Resistance, Multiple, Bacterial , Mycobacterium Infections, Nontuberculous/microbiology , Nontuberculous Mycobacteria/classification , Animals , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/pathology , Communicable Diseases, Emerging/transmission , Cystic Fibrosis/epidemiology , Cystic Fibrosis/pathology , Genome, Bacterial , Genomics , Humans , Incidence , Lung/microbiology , Lung/pathology , Mice , Mice, SCID , Mycobacterium Infections, Nontuberculous/epidemiology , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium Infections, Nontuberculous/transmission , Nontuberculous Mycobacteria/genetics , Nontuberculous Mycobacteria/isolation & purification , Phylogeny , Pneumonia, Bacterial/epidemiology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/transmission , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
10.
Nat Immunol ; 17(9): 1046-56, 2016 09.
Article in English | MEDLINE | ID: mdl-27478939

ABSTRACT

Single-nucleotide variations in C13orf31 (LACC1) that encode p.C284R and p.I254V in a protein of unknown function (called 'FAMIN' here) are associated with increased risk for systemic juvenile idiopathic arthritis, leprosy and Crohn's disease. Here we set out to identify the biological mechanism affected by these coding variations. FAMIN formed a complex with fatty acid synthase (FASN) on peroxisomes and promoted flux through de novo lipogenesis to concomitantly drive high levels of fatty-acid oxidation (FAO) and glycolysis and, consequently, ATP regeneration. FAMIN-dependent FAO controlled inflammasome activation, mitochondrial and NADPH-oxidase-dependent production of reactive oxygen species (ROS), and the bactericidal activity of macrophages. As p.I254V and p.C284R resulted in diminished function and loss of function, respectively, FAMIN determined resilience to endotoxin shock. Thus, we have identified a central regulator of the metabolic function and bioenergetic state of macrophages that is under evolutionary selection and determines the risk of inflammatory and infectious disease.


Subject(s)
Arthritis, Juvenile/genetics , Crohn Disease/genetics , Infections/genetics , Leprosy/genetics , Macrophages/immunology , Proteins/genetics , Shock, Septic/genetics , Adenosine Triphosphate/metabolism , Animals , Bacteriolysis , Cells, Cultured , Energy Metabolism , Fatty Acid Synthase, Type I/metabolism , Genetic Predisposition to Disease , Humans , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/metabolism , Oxidation-Reduction , Polymorphism, Single Nucleotide , Risk
SELECTION OF CITATIONS
SEARCH DETAIL