Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.783
1.
PLoS One ; 19(5): e0303173, 2024.
Article En | MEDLINE | ID: mdl-38739587

In this study, new series of N'-(2-(substitutedphenoxy)acetyl)-4-(1H-pyrrol-1-yl)benzohydrazides (3a-j) 4-(2,5-dimethyl-1H-pyrrol-1-yl)-N'-(2-(substitutedphenoxy)acetyl)benzohydrazides (5a-j) were synthesized, characterized and assessed as inhibitors of enoyl ACP reductase and DHFR. Most of the compounds exhibited dual inhibition against the enzymes enoyl ACP reductase and DHFR. Several synthesized substances also demonstrated significant antibacterial and antitubercular properties. A molecular docking analysis was conducted in order to determine the potential mechanism of action of the synthesized compounds. The results indicated that there were binding interactions seen with the active sites of dihydrofolate reductase and enoyl ACP reductase. Additionally, important structural details were identified that play a critical role in sustaining the dual inhibitory activity. These findings were useful for the development of future dual inhibitors. Therefore, this study provided strong evidence that several synthesized molecules could exert their antitubercular properties at the cellular level through multi-target inhibition. By shedding light on the mechanisms through which these compounds exert their inhibitory effects, this research opens up promising avenues for the future development of dual inhibitors with enhanced antibacterial and antitubercular properties. The study's findings underscore the importance of multi-target approaches in drug design, providing a strong foundation for the design and optimization of novel compounds that can effectively target bacterial infections at the cellular level.


Antitubercular Agents , Molecular Docking Simulation , Pyrroles , Tetrahydrofolate Dehydrogenase , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Antitubercular Agents/chemical synthesis , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolate Dehydrogenase/chemistry , Pyrroles/chemistry , Pyrroles/pharmacology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Microbial Sensitivity Tests , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/chemical synthesis , Humans , Structure-Activity Relationship , Catalytic Domain
2.
Cell Mol Life Sci ; 81(1): 203, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698289

Nitrogen metabolism of M. tuberculosis is critical for its survival in infected host cells. M. tuberculosis has evolved sophisticated strategies to switch between de novo synthesis and uptake of various amino acids from host cells for metabolic demands. Pyridoxal phosphate-dependent histidinol phosphate aminotransferase-HspAT enzyme is critically required for histidine biosynthesis. HspAT is involved in metabolic synthesis of histidine, phenylalanine, tyrosine, tryptophan, and novobiocin. We showed that M. tuberculosis Rv2231c is a conserved enzyme with HspAT activity. Rv2231c is a monomeric globular protein that contains α-helices and ß-sheets. It is a secretory and cell wall-localized protein that regulates critical pathogenic attributes. Rv2231c enhances the survival and virulence of recombinant M. smegmatis in infected RAW264.7 macrophage cells. Rv2231c is recognized by the TLR4 innate immune receptor and modulates the host immune response by suppressing the secretion of the antibacterial pro-inflammatory cytokines TNF, IL-12, and IL-6. It also inhibits the expression of co-stimulatory molecules CD80 and CD86 along with antigen presenting molecule MHC-I on macrophage and suppresses reactive nitrogen species formation, thereby promoting M2 macrophage polarization. Recombinant M. smegmatis expressing Rv2231c inhibited apoptosis in macrophages, promoting efficient bacterial survival and proliferation, thereby increasing virulence. Our results indicate that Rv2231c is a moonlighting protein that regulates multiple functions of M. tuberculosis pathophysiology to increase its virulence. These mechanistic insights can be used to better understand the pathogenesis of M. tuberculosis and to design strategies for tuberculosis mitigation.


Macrophages , Mycobacterium tuberculosis , Transaminases , Mice , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Animals , RAW 264.7 Cells , Virulence , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Transaminases/metabolism , Transaminases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Mycobacterium smegmatis/pathogenicity , Mycobacterium smegmatis/metabolism , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/enzymology , Cytokines/metabolism , Toll-Like Receptor 4/metabolism , Humans , Immunity, Innate , Host-Pathogen Interactions/immunology , Tuberculosis/immunology , Tuberculosis/microbiology
3.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732102

Cytochrome P450 CYP121A1 is a well-known drug target against Mycobacterium tuberculosis, the human pathogen that causes the deadly disease tuberculosis (TB). CYP121A1 is a unique P450 enzyme because it uses classical and non-classical P450 catalytic processes and has distinct structural features among P450s. However, a detailed investigation of CYP121A1 protein structures in terms of active site cavity dynamics and key amino acids interacting with bound ligands has yet to be undertaken. To address this research knowledge gap, 53 CYP121A1 crystal structures were investigated in this study. Critical amino acids required for CYP121A1's overall activity were identified and highlighted this enzyme's rigid architecture and substrate selectivity. The CYP121A1-fluconazole crystal structure revealed a novel azole drug-P450 binding mode in which azole heme coordination was facilitated by a water molecule. Fragment-based inhibitor approaches revealed that CYP121A1 can be inhibited by molecules that block the substrate channel or by directly interacting with the P450 heme. This study serves as a reference for the precise understanding of CYP121A1 interactions with different ligands and the structure-function analysis of P450 enzymes in general. Our findings provide critical information for the synthesis of more specific CYP121A1 inhibitors and their development as novel anti-TB drugs.


Cytochrome P-450 Enzyme System , Mycobacterium tuberculosis , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/drug effects , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/chemistry , Structure-Activity Relationship , Catalytic Domain , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/antagonists & inhibitors , Crystallography, X-Ray , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme Inhibitors/chemistry , Models, Molecular , Humans , Protein Binding , Substrate Specificity , Ligands , Protein Conformation
4.
ACS Infect Dis ; 10(5): 1767-1779, 2024 May 10.
Article En | MEDLINE | ID: mdl-38619138

Peptidoglycan synthesis is an underutilized drug target in Mycobacterium tuberculosis (Mtb). Diazabicyclooctanes (DBOs) are a class of broad-spectrum ß-lactamase inhibitors that also inhibit certain peptidoglycan transpeptidases that are important in mycobacterial cell wall synthesis. We evaluated the DBO durlobactam as an inhibitor of BlaC, the Mtb ß-lactamase, and multiple Mtb peptidoglycan transpeptidases (PonA1, LdtMt1, LdtMt2, LdtMt3, and LdtMt5). Timed electrospray ionization mass spectrometry (ESI-MS) captured acyl-enzyme complexes with BlaC and all transpeptidases except LdtMt5. Inhibition kinetics demonstrated durlobactam was a potent and efficient DBO inhibitor of BlaC (KI app 9.2 ± 0.9 µM, k2/K 5600 ± 560 M-1 s-1) and similar to clavulanate (KI app 3.3 ± 0.6 µM, k2/K 8400 ± 840 M-1 s-1); however, durlobactam had a lower turnover number (tn = kcat/kinact) than clavulanate (1 and 8, respectively). KI app values with durlobactam and clavulanate were similar for peptidoglycan transpeptidases, but ESI-MS captured durlobactam complexes at more time points. Molecular docking and simulation demonstrated several productive interactions of durlobactam in the active sites of BlaC, PonA1, and LdtMt2. Antibiotic susceptibility testing was conducted on 11 Mtb isolates with amoxicillin, ceftriaxone, meropenem, imipenem, clavulanate, and durlobactam. Durlobactam had a minimum inhibitory concentration (MIC) range of 0.5-16 µg/mL, similar to the ranges for meropenem (1-32 µg/mL) and imipenem (0.5-64 µg/mL). In ß-lactam + durlobactam combinations (1:1 mass/volume), MICs were lowered 4- to 64-fold for all isolates except one with meropenem-durlobactam. This work supports further exploration of novel ß-lactamase inhibitors that target BlaC and Mtb peptidoglycan transpeptidases.


Mycobacterium tuberculosis , beta-Lactamase Inhibitors , beta-Lactamases , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/metabolism , beta-Lactamases/chemistry , Peptidyl Transferases/antagonists & inhibitors , Peptidyl Transferases/metabolism , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/chemistry , Microbial Sensitivity Tests , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Molecular Docking Simulation , Peptidoglycan/metabolism , Peptidoglycan/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Kinetics , Aminoacyltransferases
5.
ACS Infect Dis ; 10(5): 1561-1575, 2024 May 10.
Article En | MEDLINE | ID: mdl-38577994

DNA-encoded chemical library (DEL) technology provides a time- and cost-efficient method to simultaneously screen billions of compounds for their affinity to a protein target of interest. Here we report its use to identify a novel chemical series of inhibitors of the thioesterase activity of polyketide synthase 13 (Pks13) from Mycobacterium tuberculosis (Mtb). We present three chemically distinct series of inhibitors along with their enzymatic and Mtb whole cell potency, the measure of on-target activity in cells, and the crystal structures of inhibitor-enzyme complexes illuminating their interactions with the active site of the enzyme. One of these inhibitors showed a favorable pharmacokinetic profile and demonstrated efficacy in an acute mouse model of tuberculosis (TB) infection. These findings and assay developments will aid in the advancement of TB drug discovery.


Antitubercular Agents , Enzyme Inhibitors , Mycobacterium tuberculosis , Small Molecule Libraries , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/drug effects , Animals , Mice , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Tuberculosis/drug therapy , Tuberculosis/microbiology , Polyketide Synthases/metabolism , Polyketide Synthases/chemistry , Polyketide Synthases/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Discovery , Drug Evaluation, Preclinical , Thiolester Hydrolases/antagonists & inhibitors , Thiolester Hydrolases/metabolism , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/genetics , Crystallography, X-Ray , Humans , Disease Models, Animal
6.
Arch Microbiol ; 206(5): 230, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38649511

During the past few decades, a wealth of knowledge has been made available for the transcription machinery in bacteria from the structural, functional and mechanistic point of view. However, comparatively little is known about the homooligomerization of the multisubunit M. tuberculosis RNA polymerase (RNAP) enzyme and its functional relevance. While E. coli RNAP has been extensively studied, many aspects of RNAP of the deadly pathogenic M. tuberculosis are still unclear. We used biophysical and biochemical methods to study the oligomerization states of the core and holoenzymes of M. tuberculosis RNAP. By size exclusion chromatography and negative staining Transmission Electron Microscopy (TEM) studies and quantitative analysis of the TEM images, we demonstrate that the in vivo reconstituted RNAP core enzyme (α2ßß'ω) can also exist as dimers in vitro. Using similar methods, we also show that the holoenzyme (core + σA) does not dimerize in vitro and exist mostly as monomers. It is tempting to suggest that the oligomeric changes that we see in presence of σA factor might have functional relevance in the cellular process. Although reported previously in E. coli, to our knowledge we report here for the first time the study of oligomeric nature of M. tuberculosis RNAP in presence and absence of σA factor.


Bacterial Proteins , DNA-Directed RNA Polymerases , Mycobacterium tuberculosis , Protein Multimerization , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Holoenzymes/chemistry , Holoenzymes/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Microscopy, Electron, Transmission , Sigma Factor/metabolism , Sigma Factor/chemistry , Sigma Factor/genetics , Chromatography, Gel
7.
ACS Infect Dis ; 10(5): 1739-1752, 2024 May 10.
Article En | MEDLINE | ID: mdl-38647213

Reverse analogs of the phosphonohydroxamic acid antibiotic fosmidomycin are potent inhibitors of the nonmevalonate isoprenoid biosynthesis enzyme 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR, IspC) of Plasmodium falciparum. Some novel analogs with large phenylalkyl substituents at the hydroxamic acid nitrogen exhibit nanomolar PfDXR inhibition and potent in vitro growth inhibition of P. falciparum parasites coupled with good parasite selectivity. X-ray crystallographic studies demonstrated that the N-phenylpropyl substituent of the newly developed lead compound 13e is accommodated in a subpocket within the DXR catalytic domain but does not reach the NADPH binding pocket of the N-terminal domain. As shown for reverse carba and thia analogs, PfDXR selectively binds the S-enantiomer of the new lead compound. In addition, some representatives of the novel inhibitor subclass are nanomolar Escherichia coli DXR inhibitors, whereas the inhibition of Mycobacterium tuberculosis DXR is considerably weaker.


Aldose-Ketose Isomerases , Antimalarials , Fosfomycin , Hydroxamic Acids , Multienzyme Complexes , Plasmodium falciparum , Fosfomycin/pharmacology , Fosfomycin/analogs & derivatives , Fosfomycin/chemistry , Aldose-Ketose Isomerases/antagonists & inhibitors , Aldose-Ketose Isomerases/metabolism , Aldose-Ketose Isomerases/chemistry , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Hydroxamic Acids/pharmacology , Hydroxamic Acids/chemistry , Antimalarials/pharmacology , Antimalarials/chemistry , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Multienzyme Complexes/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Structure-Activity Relationship , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/enzymology , Models, Molecular , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Catalytic Domain , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism
8.
Biochemistry (Mosc) ; 89(3): 407-416, 2024 Mar.
Article En | MEDLINE | ID: mdl-38648761

The synthesis of (p)ppGpp alarmones plays a vital role in the regulation of metabolism suppression, growth rate control, virulence, bacterial persistence, and biofilm formation. The (p)ppGpp alarmones are synthesized by proteins of the RelA/SpoT homolog (RSH) superfamily, including long bifunctional RSH proteins and small alarmone synthetases. Here, we investigated enzyme kinetics and dose-dependent enzyme inhibition to elucidate the mechanism of 4-(4,7-dimethyl-1,2,3,4-tetrahydronaphthalen-1-yl)pentanoic acid (DMNP) action on the (p)ppGpp synthetases RelMsm and RelZ from Mycolicibacterium smegmatis and RelMtb from Mycobacterium tuberculosis. DMNP was found to inhibit the activity of RelMtb. According to the enzyme kinetics analysis, DMNP acts as a noncompetitive inhibitor of RelMsm and RelZ. Based on the results of molecular docking, the DMNP-binding site is located in the proximity of the synthetase domain active site. This study might help in the development of alarmone synthetase inhibitors, which includes relacin and its derivatives, as well as DMNP - a synthetic analog of the marine coral metabolite erogorgiaene. Unlike conventional antibiotics, alarmone synthetase inhibitors target metabolic pathways linked to the bacterial stringent response. Although these pathways are not essential for bacteria, they regulate the development of adaptation mechanisms. Combining conventional antibiotics that target actively growing cells with compounds that impede bacterial adaptation may address challenges associated with antimicrobial resistance and bacterial persistence.


Bacterial Proteins , Ligases , Mycobacterium tuberculosis , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Kinetics , Ligases/antagonists & inhibitors , Ligases/metabolism , Molecular Docking Simulation , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/drug effects , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/drug effects , Naphthalenes/pharmacology , Naphthalenes/chemistry , Diterpenes/pharmacology
9.
Microbiol Spectr ; 12(5): e0353423, 2024 May 02.
Article En | MEDLINE | ID: mdl-38534149

To address intracellular mycobacterial infections, we developed a cocktail of four enzymes that catalytically attack three layers of the mycobacterial envelope. This cocktail is delivered to macrophages, through a targeted liposome presented here as ENTX_001. Endolytix Cocktail 1 (EC1) leverages mycobacteriophage lysin enzymes LysA and LysB, while also including α-amylase and isoamylase for degradation of the mycobacterial envelope from outside of the cell. The LysA family of proteins from mycobacteriophages has been shown to cleave the peptidoglycan layer, whereas LysB is an esterase that hydrolyzes the linkage between arabinogalactan and mycolic acids of the mycomembrane. The challenge of gaining access to the substrates of LysA and LysB provided exogenously was addressed by adding amylase enzymes that degrade the extracellular capsule shown to be present in Mycobacterium tuberculosis. This enzybiotic approach avoids antimicrobial resistance, specific receptor-mediated binding, and intracellular DNA surveillance pathways that limit many bacteriophage applications. We show this cocktail of enzymes is bactericidal in vitro against both rapid- and slow-growing nontuberculous mycobacteria (NTM) as well as M. tuberculosis strains. The EC1 cocktail shows superior killing activity when compared to previously characterized LysB alone. EC1 is also powerfully synergistic with standard-of-care antibiotics. In addition to in vitro killing of NTM, ENTX_001 demonstrates the rescue of infected macrophages from necrotic death by Mycobacteroides abscessus and Mycobacterium avium. Here, we demonstrate shredding of mycobacterial cells by EC1 into cellular debris as a mechanism of bactericide.IMPORTANCEThe world needs entirely new forms of antibiotics as resistance to chemical antibiotics is a critical problem facing society. We addressed this need by developing a targeted enzyme therapy for a broad range of species and strains within mycobacteria and highly related genera including nontuberculous mycobacteria such as Mycobacteroides abscessus, Mycobacterium avium, Mycobacterium intracellulare, as well as Mycobacterium tuberculosis. One advantage of this approach is the ability to drive our lytic enzymes through encapsulation into macrophage-targeted liposomes resulting in attack of mycobacteria in the cells that harbor them where they hide from the adaptive immune system and grow. Furthermore, this approach shreds mycobacteria independent of cell physiology as the drug targets the mycobacterial envelope while sidestepping the host range limitations observed with phage therapy and resistance to chemical antibiotics.


Galactans , Macrophages , Mycobacteriophages , Mycobacterium tuberculosis , Nontuberculous Mycobacteria , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Mycobacteriophages/genetics , Mycobacteriophages/enzymology , Macrophages/microbiology , Macrophages/virology , Humans , Nontuberculous Mycobacteria/drug effects , Liposomes/chemistry , Anti-Bacterial Agents/pharmacology , Peptidoglycan/metabolism , Microbial Sensitivity Tests , Endopeptidases/metabolism , Endopeptidases/pharmacology , Endopeptidases/genetics
10.
Nature ; 628(8006): 186-194, 2024 Apr.
Article En | MEDLINE | ID: mdl-38509362

Drug-resistant bacteria are emerging as a global threat, despite frequently being less fit than their drug-susceptible ancestors1-8. Here we sought to define the mechanisms that drive or buffer the fitness cost of rifampicin resistance (RifR) in the bacterial pathogen Mycobacterium tuberculosis (Mtb). Rifampicin inhibits RNA polymerase (RNAP) and is a cornerstone of modern short-course tuberculosis therapy9,10. However, RifR Mtb accounts for one-quarter of all deaths due to drug-resistant bacteria11,12. We took a comparative functional genomics approach to define processes that are differentially vulnerable to CRISPR interference (CRISPRi) inhibition in RifR Mtb. Among other hits, we found that the universally conserved transcription factor NusG is crucial for the fitness of RifR Mtb. In contrast to its role in Escherichia coli, Mtb NusG has an essential RNAP pro-pausing function mediated by distinct contacts with RNAP and the DNA13. We find this pro-pausing NusG-RNAP interface to be under positive selection in clinical RifR Mtb isolates. Mutations in the NusG-RNAP interface reduce pro-pausing activity and increase fitness of RifR Mtb. Collectively, these results define excessive RNAP pausing as a molecular mechanism that drives the fitness cost of RifR in Mtb, identify a new mechanism of compensation to overcome this cost, suggest rational approaches to exacerbate the fitness cost, and, more broadly, could inform new therapeutic approaches to develop drug combinations to slow the evolution of RifR in Mtb.


Bacterial Proteins , Drug Resistance, Bacterial , Evolution, Molecular , Genetic Fitness , Mycobacterium tuberculosis , Rifampin , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Conserved Sequence , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Genomics , Mutation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Rifampin/pharmacology , Rifampin/therapeutic use , Transcription Factors/genetics , Transcription Factors/metabolism , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/microbiology
11.
J Mol Biol ; 436(2): 168379, 2024 01 15.
Article En | MEDLINE | ID: mdl-38043732

Phosphosignaling in bacteria is mediated by two distinct systems, the two-component systems (TCSs) and the protein Ser/Thr/Tyr, or O-phosphorylation systems. These two arms of phosphosignaling are currently thought to be largely independent from one another. We mined a deep Mycobacterium tuberculosis (Mtb) phosphoproteome and identified over 170 O-phosphorylation sites on histidine kinases and response regulators of TCSs, suggesting that the two signaling pathways extensively intersect. Several TCSs were phosphorylated on multiple sites, and many by multiple Ser/Thr protein kinases, suggesting convergent and cooperative regulatory interactions. To test in which way these O-phosphorylation sites affect TCS activity, we reconstituted the NarSL phosphorelay in vitro. The Ser/Thr protein kinase PknL phosphorylated the histidine kinase NarS and activated its autophosphorylating activity. A phosphoablative mutation at the PknL phosphorylation site Thr380 resulted in low autophosphorylating activity, whereas a phosphomimetic mutation strongly activated autophosphorylation. The phosphomimetic mutation also resulted in more efficient phosphotransfer from NarS to the response regulator NarL and suppression of gene expression. These data show control of NarSL signaling by STPKs through a phosphoswitch and point to extensive, functional crosstalk between TCSs and O-phosphorylation.


Bacterial Proteins , Mycobacterium tuberculosis , Protein Serine-Threonine Kinases , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
12.
J Biol Chem ; 299(7): 104924, 2023 07.
Article En | MEDLINE | ID: mdl-37328106

Mycobacterium tuberculosis's (Mtb) success as a pathogen is due in part to its sophisticated lipid metabolic programs, both catabolic and biosynthetic. Several of Mtb lipids have specific roles in pathogenesis, but the identity and roles of many are unknown. Here, we demonstrated that the tyz gene cluster in Mtb, previously implicated in resistance to oxidative stress and survival in macrophages, encodes the biosynthesis of acyl-oxazolones. Heterologous expression of tyzA (Rv2336), tyzB (Rv2338c) and tyzC (Rv2337c) resulted in the biosynthesis of C12:0-tyrazolone as the predominant compound, and the C12:0-tyrazolone was identified in Mtb lipid extracts. TyzA catalyzed the N-acylation of l-amino acids, with highest specificity for l-Tyr and l-Phe and lauroyl-CoA (kcat/KM = 5.9 ± 0.8 × 103 M-1s-1). In cell extracts, TyzC, a flavin-dependent oxidase (FDO) of the nitroreductase (NTR) superfamily, catalyzed the O2-dependent desaturation of the N-acyl-L-Tyr produced by TyzA, while TyzB, a ThiF homolog, catalyzed its ATP-dependent cyclization. The substrate preference of TyzB and TyzC appear to determine the identity of the acyl-oxazolone. Phylogenetic analyses revealed that the NTR superfamily includes a large number of broadly distributed FDOs, including five in Mtb that likely catalyze the desaturation of lipid species. Finally, TCA1, a molecule with activity against drug-resistant and persistent tuberculosis, failed to inhibit the cyclization activity of TyzB, the proposed secondary target of TCA1. Overall, this study identifies a novel class of Mtb lipids, clarifies the role of a potential drug target, and expands our understanding of the NTR superfamily.


Lipids , Mycobacterium tuberculosis , Nitroreductases , Lipids/biosynthesis , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Phylogeny
13.
Nat Commun ; 14(1): 3828, 2023 06 28.
Article En | MEDLINE | ID: mdl-37380634

Mycobacterium tuberculosis is one of the global leading causes of death due to a single infectious agent. Pretomanid and delamanid are new antitubercular agents that have progressed through the drug discovery pipeline. These compounds are bicyclic nitroimidazoles that act as pro-drugs, requiring activation by a mycobacterial enzyme; however, the precise mechanisms of action of the active metabolite(s) are unclear. Here, we identify a molecular target of activated pretomanid and delamanid: the DprE2 subunit of decaprenylphosphoribose-2'-epimerase, an enzyme required for the synthesis of cell wall arabinogalactan. We also provide evidence for an NAD-adduct as the active metabolite of pretomanid. Our results highlight DprE2 as a potential antimycobacterial target and provide a foundation for future exploration into the active metabolites and clinical development of pretomanid and delamanid.


Antitubercular Agents , Mycobacterium tuberculosis , Nitroimidazoles , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Molecular Targeted Therapy , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Alcohol Oxidoreductases/antagonists & inhibitors , Nitroimidazoles/pharmacology , Nitroimidazoles/therapeutic use , Cell Wall/metabolism , Drug Resistance , Prodrugs/chemistry , Prodrugs/metabolism , Spectrophotometry , NAD/metabolism , Kinetics
14.
J Biol Chem ; 299(5): 103003, 2023 05.
Article En | MEDLINE | ID: mdl-36775125

DNA gyrase is an essential nucleoprotein motor present in all bacteria and is a major target for antibiotic treatment of Mycobacterium tuberculosis (MTB) infection. Gyrase hydrolyzes ATP to add negative supercoils to DNA using a strand passage mechanism that has been investigated using biophysical and biochemical approaches. To analyze the dynamics of substeps leading to strand passage, single-molecule rotor bead tracking (RBT) has been used previously to follow real-time supercoiling and conformational transitions in Escherichia coli (EC) gyrase. However, RBT has not yet been applied to gyrase from other pathogenically relevant bacteria, and it is not known whether substeps are conserved across evolutionarily distant species. Here, we compare gyrase supercoiling dynamics between two evolutionarily distant bacterial species, MTB and EC. We used RBT to measure supercoiling rates, processivities, and the geometries and transition kinetics of conformational states of purified gyrase proteins in complex with DNA. Our results show that E. coli and MTB gyrases are both processive, with the MTB enzyme displaying velocities ∼5.5× slower than the EC enzyme. Compared with EC gyrase, MTB gyrase also more readily populates an intermediate state with DNA chirally wrapped around the enzyme, in both the presence and absence of ATP. Our substep measurements reveal common features in conformational states of EC and MTB gyrases interacting with DNA but also suggest differences in populations and transition rates that may reflect distinct cellular needs between these two species.


DNA Gyrase , Escherichia coli , Mycobacterium tuberculosis , Adenosine Triphosphate/metabolism , DNA , DNA Gyrase/chemistry , DNA Gyrase/metabolism , DNA, Superhelical , Escherichia coli/enzymology , Escherichia coli/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Molecular Dynamics Simulation
15.
Biochemistry ; 62(3): 710-721, 2023 02 07.
Article En | MEDLINE | ID: mdl-36657084

Over one and a half million people die of tuberculosis (TB) each year. Multidrug-resistant TB infections are especially dangerous, and new drugs are needed to combat them. The high cost and complexity of drug development make repositioning of drugs that are already in clinical use for other indications a potentially time- and money-saving avenue. In this study, we identified among existing drugs five compounds: azelastine, venlafaxine, chloroquine, mefloquine, and proguanil as inhibitors of acetyltransferase Eis from Mycobacterium tuberculosis, a causative agent of TB. Eis upregulation is a cause of clinically relevant resistance of TB to kanamycin, which is inactivated by Eis-catalyzed acetylation. Crystal structures of these drugs as well as chlorhexidine in complexes with Eis showed that these inhibitors were bound in the aminoglycoside binding cavity, consistent with their established modes of inhibition with respect to kanamycin. Among three additionally synthesized compounds, a proguanil analogue, designed based on the crystal structure of the Eis-proguanil complex, was 3-fold more potent than proguanil. The crystal structures of these compounds in complexes with Eis explained their inhibitory potencies. These initial efforts in rational drug repositioning can serve as a starting point in further development of Eis inhibitors.


Acetyltransferases , Mycobacterium tuberculosis , Tuberculosis , Humans , Acetyltransferases/antagonists & inhibitors , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Kanamycin/pharmacology , Kanamycin/chemistry , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Proguanil/metabolism , Tuberculosis/drug therapy
16.
Nucleic Acids Res ; 51(1): 349-364, 2023 01 11.
Article En | MEDLINE | ID: mdl-36583363

Each catalytic cycle of type IA topoisomerases has been proposed to comprise multistep reactions. The capture of the transport-segment DNA (T-segment) into the central cavity of the N-terminal toroidal structure is an important action, which is preceded by transient gate-segment (G-segment) cleavage and succeeded by G-segment religation for the relaxation of negatively supercoiled DNA and decatenation of DNA. The T-segment passage in and out of the central cavity requires significant domain-domain rearrangements, including the movement of D3 relative to D1 and D4 for the opening and closing of the gate towards the central cavity. Here we report a direct observation of the interaction of a duplex DNA in the central cavity of a type IA topoisomerase and its associated domain-domain conformational changes in a crystal structure of a Mycobacterium tuberculosis topoisomerase I complex that also has a bound G-segment. The duplex DNA within the central cavity illustrates the non-sequence-specific interplay between the T-segment DNA and the enzyme. The rich structural information revealed from the novel topoisomerase-DNA complex, in combination with targeted mutagenesis studies, provides new insights into the mechanism of the topoisomerase IA catalytic cycle.


DNA Topoisomerases, Type I , DNA , Mycobacterium tuberculosis , DNA/chemistry , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Models, Molecular , Mycobacterium tuberculosis/enzymology
17.
Eur J Med Chem ; 245(Pt 1): 114908, 2023 Jan 05.
Article En | MEDLINE | ID: mdl-36435016

Herein a series of 4-aminoquinolines were synthesized in an attempt to optimize and study the structural features related to LABIO-17 biological activity, a Mycobacterium tuberculosis NADH-dependent enoyl-acyl carrier protein reductase (MtInhA) inhibitor previously identified by a virtual-ligand-screening approach. Structure-activity relationships led to novel submicromolar inhibitors of MtInhA and potent antitubercular agents. The lead compound is 87-fold more potent as enzymatic inhibitors and 32-fold more potent against M. tuberculosis H37Rv strain in comparison with LABIO-17. These molecules were also active against multidrug-resistant strains, devoid of apparent toxicity to mammalian cells and showed favorable in vitro ADME profiles. Additionally, these compounds were active in an intracellular model of tuberculosis (TB) infection, showed no genotoxicity signals, satisfactory absorption parameters and absence of in vivo acute toxicity. Finally, treatment with selected 4-aminoquinoline for two weeks produced bacteriostatic effect in a murine model of TB. Taken together, these findings indicate that this chemical class may furnish candidates for the future development of drug-sensitive and drug-resistant tuberculosis treatments.


Aminoquinolines , Antitubercular Agents , Enzyme Inhibitors , Mycobacterium tuberculosis , NAD (+) and NADP (+) Dependent Alcohol Oxidoreductases , Animals , Mice , Aminoquinolines/chemical synthesis , Aminoquinolines/pharmacology , Aminoquinolines/therapeutic use , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , NAD (+) and NADP (+) Dependent Alcohol Oxidoreductases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Tuberculosis/drug therapy , Disease Models, Animal
18.
Eur J Med Chem ; 245(Pt 1): 114896, 2023 Jan 05.
Article En | MEDLINE | ID: mdl-36370551

The cytochrome bcc-aa3 oxidase (Cyt-bcc) of Mycobacterium tuberculosis (Mtb) is a promising anti-tuberculosis target. However, when Cyt-bcc is inhibited, cytochrome bd terminal oxidase (Cyt-bd) can still maintain the activity of the respiratory chain and drive ATP synthesis. Through virtual screening and biological validation, we discovered two FDA-approved drugs, ivacaftor and roquinimex, exhibited moderate binding affinity to Cyt-bd. Structural modifications of them led to 1-hydroxy-2-methylquinolin-4(1H)-one derivatives as potent new Cyt-bd inhibitors. Compound 8d binds to Cyt-bd with a Kd value of 4.17 µM and inhibits the growth of the Cyt-bcc knock-out strain (ΔqcrCAB, Cyt-bd+) with a MIC value of 6.25 µM. The combination of 8d with the Cyt-bcc inhibitor Q203 completely inhibited oxygen consumption of the wild-type strain and the inverted-membrane vesicles expressing M. tuberculosis Cyt-bd (ΔcydAB::MtbCydAB+). Our study provides a promising starting point for the development of novel dual chemotherapies for tuberculosis.


Antitubercular Agents , Cytochrome b Group , Cytochrome d Group , Mycobacterium tuberculosis , Oxidoreductases , Humans , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Oxidoreductases/antagonists & inhibitors , Tuberculosis/drug therapy , Cytochrome b Group/antagonists & inhibitors , Cytochrome d Group/antagonists & inhibitors
19.
FEBS J ; 290(6): 1563-1582, 2023 03.
Article En | MEDLINE | ID: mdl-36197115

A hallmark of Mycobacterium tuberculosis (M. tb), the aetiologic agent of tuberculosis, is its ability to metabolise host-derived lipids. However, the enzymes and mechanisms underlying such metabolism are still largely unknown. We previously reported that the Cyclophostin & Cyclipostins (CyC) analogues, a new family of potent antimycobacterial molecules, react specifically and covalently with (Ser/Cys)-based enzymes mostly involved in bacterial lipid metabolism. Here, we report the synthesis of new CyC alkyne-containing inhibitors (CyCyne ) and their use for the direct fishing of target proteins in M. tb culture via bio-orthogonal click-chemistry activity-based protein profiling (CC-ABPP). This approach led to the capture and identification of a variety of enzymes, and many of them involved in lipid or steroid metabolisms. One of the captured enzymes, HsaD (Rv3569c), is required for the survival of M. tb within macrophages and is thus a potential therapeutic target. This prompted us to further explore and validate, through a combination of biochemical and structural approaches, the specificity of HsaD inhibition by the CyC analogues. We confirmed that the CyC bind covalently to the catalytic Ser114 residue, leading to a total loss of enzyme activity. These data were supported by the X-ray structures of four HsaD-CyC complexes, obtained at resolutions between 1.6 and 2.6 Å. The identification of mycobacterial enzymes directly captured by the CyCyne probes through CC-ABPP paves the way to better understand and potentially target key players at crucial stages of the bacilli life cycle.


Antitubercular Agents , Bacterial Proteins , Hydrolases , Molecular Docking Simulation , Mycobacterium tuberculosis , Organophosphorus Compounds , Humans , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Macrophages/microbiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Tuberculosis/drug therapy , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Organophosphorus Compounds/chemistry , Crystallography, X-Ray , Hydrolases/antagonists & inhibitors , Hydrolases/chemistry , Computer Simulation
20.
ACS Infect Dis ; 8(12): 2430-2440, 2022 12 09.
Article En | MEDLINE | ID: mdl-36417754

Menaquinone (MK) is an essential component of the electron transport chain (ETC) in the gram-variable Mycobacterium tuberculosis and many Gram-positive pathogens. Three genes in the M. tuberculosis genome were annotated as methyltransferases involved in lipoquinone synthesis in mycobacteria. Heterologous expression of Rv0558 complemented an ubiE (the quinone C-methyltransferase involved in ubiquinone and menaquinone synthesis) deletion in Escherichia coli, and expression in a wild-type E. coli strain increased quinone C-methyltransferase specific activity by threefold. Rv0558 encodes a canonical C-methyltransferase or, more specifically, a S-adenosylmethionine/demethylmenaquinol methyltransferase. Partially purified recombinant protein catalyzed the formation of MK from demethylmenaquinone (DMK), although the activity of the recombinant protein was low and appeared to require a cofactor or intact membrane structure for activity. Membrane preparations from irradiated M. tuberculosis also showed poor activity; however, membrane preparations from wild-type Mycobacterium smegmatis showed robust, substrate-dependent activity. The apparent Km values for demethylmenaquinone and SAM were 14 ± 5.0 and 17 ± 7.0 µM, respectively. Interestingly, addition of dithiothreitol, dithionite, NADH, or other substrates of primary dehydrogenases to reaction mixtures containing membrane preparations stimulated the activity. Thus, these observations strongly suggest that demethylmenaquinol is the actual substrate of MenG. Ro 48-8071, previously reported to inhibit mycobacterial MK synthesis and growth, inhibited Rv0558 activity with an IC50 value of 5.1 ± 0.5 µM, and DG70 (GSK1733953A), first described as a respiration inhibitor in M. tuberculosis, inhibits MenG activity with an IC50 value of 2.6 ± 0.6 µM.


Bacterial Proteins , Methyltransferases , Mycobacterium tuberculosis , Vitamin K 2 , Humans , Escherichia coli/genetics , Methyltransferases/antagonists & inhibitors , Methyltransferases/chemistry , Methyltransferases/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Vitamin K 2/metabolism
...