Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
FEMS Microbes ; 5: xtae015, 2024.
Article in English | MEDLINE | ID: mdl-38813097

ABSTRACT

Enterococcus faecium is a gut commensal bacterium which is gaining increasing relevance as an opportunistic, nosocomial pathogen. Its high level of intrinsic and acquired antimicrobial resistance is causing a lack of treatment options, particularly for infections with vancomycin-resistant strains, and prioritizes the identification and functional validation of novel druggable targets. Here, we use activity-based protein profiling (ABPP), a chemoproteomics approach using functionalized covalent inhibitors, to detect active serine hydrolases across 11 E. faecium and Enterococcus lactis strains. Serine hydrolases are a big and diverse enzyme family, that includes known drug targets such as penicillin-binding proteins (PBPs), whereas other subfamilies are underexplored. Comparative gel-based ABPP using Bocillin-FL revealed strain- and growth condition-dependent variations in PBP activities. Profiling with the broadly serine hydrolase-reactive fluorescent probe fluorophosphonate-TMR showed a high similarity across E. faecium clade A1 strains, but higher variation across A2 and E. lactis strains. To identify these serine hydrolases, we used a biotinylated probe analog allowing for enrichment and identification via liquid chromatography-mass spectrometry. We identified 11 largely uncharacterized targets (α,ß-hydrolases, SGNH-hydrolases, phospholipases, and amidases, peptidases) that are druggable and accessible in live vancomycin-resistant E. faecium E745 and may possess vital functions that are to be characterized in future studies.

2.
mSystems ; 9(5): e0017924, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38656122

ABSTRACT

The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE: This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.


Subject(s)
Adenosine Triphosphate , Host-Pathogen Interactions , Keratinocytes , Macrophages , Staphylococcus aureus , Humans , Staphylococcus aureus/metabolism , Adenosine Triphosphate/metabolism , Host-Pathogen Interactions/immunology , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Keratinocytes/microbiology , Keratinocytes/metabolism , Keratinocytes/immunology , THP-1 Cells , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Proteomics/methods , Bacterial Proteins/metabolism , HaCaT Cells
3.
mSystems ; 9(4): e0013024, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38470253

ABSTRACT

In the Staphylococcus aureus genome, a set of highly conserved two-component systems (TCSs) composed of histidine kinases (HKs) and their cognate response regulators (RRs) sense and respond to environmental stimuli, which drive the adaptation of the bacteria. This study investigates the complex interplay between TCSs in S. aureus USA300, a predominant methicillin-resistant S. aureus strain, revealing shared and unique virulence regulatory pathways and genetic variations mediating signal specificity within TCSs. Using TCS-related mutants from the Nebraska Transposon Mutant Library, we analyzed the effects of inactivated TCS HKs and RRs on the production of various virulence factors, in vitro infection abilities, and adhesion assays. We found that the TCSs' influence on virulence determinants was not associated with their phylogenetic relationship, indicating divergent functional evolution. Using the co-crystallized structure of the DesK-DesR from Bacillus subtilis and the modeled structures of the four NarL TCSs in S. aureus, we identified interacting residues, revealing specificity determinants and conservation within the same TCS, even from different strain backgrounds. The interacting residues were highly conserved within strains but varied between species due to selection pressures and the coevolution of cognate pairs. This study unveils the complex interplay and divergent functional evolution of TCSs, highlighting their potential for future experimental exploration of phosphotransfer between cognate and non-cognate recombinant HK and RRs.IMPORTANCEGiven the widespread conservation of two-component systems (TCSs) in bacteria and their pivotal role in regulating metabolic and virulence pathways, they present a compelling target for anti-microbial agents, especially in the face of rising multi-drug-resistant infections. Harnessing TCSs therapeutically necessitates a profound understanding of their evolutionary trajectory in signal transduction, as this underlies their unique or shared virulence regulatory pathways. Such insights are critical for effectively targeting TCS components, ensuring an optimized impact on bacterial virulence, and mitigating the risk of resistance emergence via the evolution of alternative pathways. Our research offers an in-depth exploration of virulence determinants controlled by TCSs in S. aureus, shedding light on the evolving specificity determinants that orchestrate interactions between their cognate pairs.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Virulence/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Phylogeny , Bacteria/metabolism
4.
ACS Infect Dis ; 9(11): 2119-2132, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37824340

ABSTRACT

The development of new treatment options for bacterial infections requires access to new targets for antibiotics and antivirulence strategies. Chemoproteomic approaches are powerful tools for profiling and identifying novel druggable target candidates, but their functions often remain uncharacterized. Previously, we used activity-based protein profiling in the opportunistic pathogen Staphylococcus aureus to identify active serine hydrolases termed fluorophosphonate-binding hydrolases (Fph). Here, we provide the first characterization of S. aureus FphH, a conserved, putative carboxylesterase (referred to as yvaK in Bacillus subtilis) at the molecular and cellular level. First, phenotypic characterization of fphH-deficient transposon mutants revealed phenotypes during growth under nutrient deprivation, biofilm formation, and intracellular survival. Biochemical and structural investigations revealed that FphH acts as an esterase and lipase based on a fold well suited to act on a small to long hydrophobic unbranched lipid group within its substrate and can be inhibited by active site-targeting oxadiazoles. Prompted by a previous observation that fphH expression was upregulated in response to fusidic acid, we found that FphH can deacetylate this ribosome-targeting antibiotic, but the lack of FphH function did not infer major changes in antibiotic susceptibility. In conclusion, our results indicate a functional role of this hydrolase in S. aureus stress responses, and hypothetical functions connecting FphH with components of the ribosome rescue system that are conserved in the same gene cluster across Bacillales are discussed. Our atomic characterization of FphH will facilitate the development of specific FphH inhibitors and probes to elucidate its physiological role and validity as a drug target.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Fusidic Acid , Endopeptidases/metabolism , Staphylococcal Infections/microbiology
5.
Chembiochem ; 24(21): e202300473, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37552008

ABSTRACT

Activity-based protein profiling is a powerful chemoproteomic technique to detect active enzymes and identify targets and off-targets of drugs. Here, we report the use of carmofur- and activity-based probes to identify biologically relevant enzymes in the bacterial pathogen Staphylococcus aureus. Carmofur is an anti-neoplastic prodrug of 5-fluorouracil and also has antimicrobial and anti-biofilm activity. Carmofur probes were originally designed to target human acid ceramidase, a member of the NTN hydrolase family with an active-site cysteine nucleophile. Here, we first profiled the targets of a fluorescent carmofur probe in live S. aureus under biofilm-promoting conditions and in liquid culture, before proceeding to target identification by liquid chromatography/mass spectrometry. Treatment with a carmofur-biotin probe led to enrichment of 20 enzymes from diverse families awaiting further characterization, including the NTN hydrolase-related IMP cyclohydrolase PurH. However, the probe preferentially labeled serine hydrolases, thus displaying a reactivity profile similar to that of carbamates. Our results suggest that the electrophilic N-carbamoyl-5-fluorouracil scaffold could potentially be optimized to achieve selectivity towards diverse enzyme families. The observed promiscuous reactivity profile suggests that the clinical use of carmofur presumably leads to inactivation of a number human and microbial enzymes, which could lead to side effects and/or contribute to therapeutic efficacy.


Subject(s)
Anti-Infective Agents , Methicillin-Resistant Staphylococcus aureus , Humans , Staphylococcus aureus , Fluorouracil/chemistry , Fluorouracil/pharmacology , Anti-Infective Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Biofilms
6.
Molecules ; 25(21)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114655

ABSTRACT

Chemical probes have been instrumental in microbiology since its birth as a discipline in the 19th century when chemical dyes were used to visualize structural features of bacterial cells for the first time. In this review article we will illustrate the evolving design of chemical probes in modern chemical biology and their diverse applications in bacterial imaging and phenotypic analysis. We will introduce and discuss a variety of different probe types including fluorogenic substrates and activity-based probes that visualize metabolic and specific enzyme activities, metabolic labeling strategies to visualize structural features of bacterial cells, antibiotic-based probes as well as fluorescent conjugates to probe biomolecular uptake pathways.


Subject(s)
Bacteria/chemistry , Bacteria/cytology , Bacterial Physiological Phenomena , Microbiological Techniques/methods , Molecular Probes/chemistry , Staining and Labeling
7.
ACS Infect Dis ; 6(10): 2771-2782, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32865965

ABSTRACT

Staphylococcus aureus is a prevalent bacterial pathogen in both community and hospital settings, and its treatment is made particularly difficult by resilience within biofilms. Within this niche, serine hydrolase enzymes play a key role in generating and maintaining the biofilm matrix. Activity-based profiling has previously identified a family of serine hydrolases, designated fluorophosphonate-binding hydrolases (Fph's), some of which contribute to the virulence of S. aureus in vivo. These 10 Fph proteins have limited annotation and have few, if any, characterized bacterial or mammalian homologues. This suggests unique hydrolase functions even within bacterial species. Here we report structures of one of the most abundant Fph family members, FphF. Our structures capture FphF alone, covalently bound to a substrate analogue and bound to small molecule inhibitors that occupy the hydrophobic substrate-binding pocket. In line with these findings, we show that FphF has promiscuous esterase activity toward hydrophobic lipid substrates. We present docking studies that characterize interactions of inhibitors and substrates within the active site environment, which can be extended to other Fph family members. Comparison of FphF to other esterases and the wider Fph protein family suggest that FphF forms a new esterase subfamily. Our data suggest that other Fph enzymes, including the virulence factor FphB, are likely to have more restricted substrate profiles than FphF. This work demonstrates a clear molecular rationale for the specificity of fluorophosphonate probes that target FphF and provides a structural template for the design of enhanced probes and inhibitors of the Fph family of serine hydrolases.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Hydrolases/genetics , Hydrolases/metabolism , Serine , Staphylococcus aureus/metabolism , Substrate Specificity
8.
ACS Infect Dis ; 6(5): 930-938, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32298574

ABSTRACT

The bacterial genus Staphylococcus comprises diverse species that colonize the skin as commensals but can also cause infection. Previous work identified a family of serine hydrolases termed fluorophoshonate-binding hydrolases (Fphs) in the pathogenic bacteria Staphylococcus aureus, one of which, FphB, functions as a virulence factor. Using a combination of bioinformatics and activity-based protein profiling (ABPP), we identify homologues of these enzymes in the related commensal bacteria Staphylococcus epidermidis. Two of the S. aureus Fph enzymes were not identified in S. epidermidis. Using ABPP, we identified several candidate hydrolases that were not previously identified in S. aureus that may be functionally related to the Fphs. Interestingly, the activity of the Fphs vary across clinical isolates of S. epidermidis. Biochemical characterization of the FphB homologue in S. epidermidis (SeFphB) suggests it is a functional homologue of FphB in S. aureus, but our preliminary studies suggest it may not have a role in colonization in vivo. This potential difference in biological function between the Fphs of closely related staphylococcal species may provide mechanisms for specific inhibition of S. aureus infection without perturbing commensal communities of related bacteria.


Subject(s)
Bacterial Proteins/metabolism , Hydrolases/metabolism , Staphylococcus epidermidis , Virulence Factors/metabolism , Bacterial Proteins/genetics , Humans , Hydrolases/genetics , Serine , Skin/microbiology , Staphylococcal Infections , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/genetics , Virulence Factors/genetics
9.
Biol Chem ; 401(2): 233-248, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31939273

ABSTRACT

Molecular imaging methods can provide spatio-temporal information about the distribution of biomolecules or biological processes, such as certain enzymatic activities, in single cells. Within a cell, it is possible to define the subcellular location of a target, its trafficking through the cell, colocalization with other biomolecules of interest and involvement in certain cell biological processes. On the other hand, single-cell imaging promises to distinguish cells that are phenotypically different from each other. The corresponding cellular diversity comprises the presence of functionally distinct cells in a population ('phenotypic heterogeneity'), as well as dynamic cellular responses to external stimuli ('phenotypic plasticity'), which is highly relevant, e.g. during cell differentiation, activation (of immune cells), or cell death. This review focuses on applications of a certain class of chemical probes, the so-called activity-based probes (ABPs), for visualization of enzymatic activities in the single-cell context. It discusses the structure of ABPs and other chemical probes, exemplary applications of ABPs in single-cell studies in human, mouse and bacterial systems and considerations to be made with regard to data interpretation.


Subject(s)
Enzymes/analysis , Enzymes/metabolism , Molecular Imaging , Single-Cell Analysis , Animals , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Humans
10.
Angew Chem Int Ed Engl ; 58(17): 5643-5647, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30768830

ABSTRACT

Phenotypically distinct cellular (sub)populations are clinically relevant for the virulence and antibiotic resistance of a bacterial pathogen, but functionally different cells are usually indistinguishable from each other. Herein, we introduce fluorescent activity-based probes as chemical tools for the single-cell phenotypic characterization of enzyme activity levels in Staphylococcus aureus. We screened a 1,2,3-triazole urea library to identify selective inhibitors of fluorophosphonate-binding serine hydrolases and lipases in S. aureus and synthesized target-selective activity-based probes. Molecular imaging and activity-based protein profiling studies with these probes revealed a dynamic network within this enzyme family involving compensatory regulation of specific family members and exposed single-cell phenotypic heterogeneity. We propose the labeling of enzymatic activities by chemical probes as a generalizable method for the phenotyping of bacterial cells at the population and single-cell level.


Subject(s)
Fluorescent Dyes/metabolism , Staphylococcus aureus/metabolism , Triazoles/metabolism , Urea/metabolism , Humans , Phenotype
11.
Nat Chem Biol ; 14(6): 609-617, 2018 06.
Article in English | MEDLINE | ID: mdl-29769740

ABSTRACT

Serine hydrolases play diverse roles in regulating host-pathogen interactions in a number of organisms, yet few have been characterized in the human pathogen Staphylococcus aureus. Here we describe a chemical proteomic screen that identified ten previously uncharacterized S. aureus serine hydrolases that mostly lack human homologs. We termed these enzymes fluorophosphonate-binding hydrolases (FphA-J). One hydrolase, FphB, can process short fatty acid esters, exhibits increased activity in response to host cell factors, is located predominantly on the bacterial cell surface in a subset of cells, and is concentrated in the division septum. Genetic disruption of fphB confirmed that the enzyme is dispensable for bacterial growth in culture but crucial for establishing infection in distinct sites in vivo. A selective small molecule inhibitor of FphB effectively reduced infectivity in vivo, suggesting that it may be a viable therapeutic target for the treatment or management of Staphylococcus infections.


Subject(s)
Bacterial Proteins/metabolism , Hydrolases/metabolism , Staphylococcus aureus/metabolism , Virulence Factors/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Binding Sites , Cloning, Molecular , Fatty Acids/chemistry , Genetic Techniques , HEK293 Cells , Host-Pathogen Interactions , Humans , Hydrolysis , Kinetics , Mice , Microbial Sensitivity Tests , Organophosphonates/chemistry , Phylogeny , Proteomics/methods , Serine/chemistry , Staphylococcal Infections , Virulence , Virulence Factors/genetics
12.
ACS Cent Sci ; 3(11): 1143-1155, 2017 Nov 22.
Article in English | MEDLINE | ID: mdl-29202016

ABSTRACT

Proteasome inhibitors are used to treat blood cancers such as multiple myeloma (MM) and mantle cell lymphoma. The efficacy of these drugs is frequently undermined by acquired resistance. One mechanism of proteasome inhibitor resistance may involve the transcription factor Nuclear Factor, Erythroid 2 Like 1 (NFE2L1, also referred to as Nrf1), which responds to proteasome insufficiency or pharmacological inhibition by upregulating proteasome subunit gene expression. This "bounce-back" response is achieved through a unique mechanism. Nrf1 is constitutively translocated into the ER lumen, N-glycosylated, and then targeted for proteasomal degradation via the ER-associated degradation (ERAD) pathway. Proteasome inhibition leads to accumulation of cytosolic Nrf1, which is then processed to form the active transcription factor. Here we show that the cytosolic enzyme N-glycanase 1 (NGLY1, the human PNGase) is essential for Nrf1 activation in response to proteasome inhibition. Chemical or genetic disruption of NGLY1 activity results in the accumulation of misprocessed Nrf1 that is largely excluded from the nucleus. Under these conditions, Nrf1 is inactive in regulating proteasome subunit gene expression in response to proteasome inhibition. Through a small molecule screen, we identified a cell-active NGLY1 inhibitor that disrupts the processing and function of Nrf1. The compound potentiates the cytotoxicity of carfilzomib, a clinically used proteasome inhibitor, against MM and T cell-derived acute lymphoblastic leukemia (T-ALL) cell lines. Thus, NGLY1 inhibition prevents Nrf1 activation and represents a new therapeutic approach for cancers that depend on proteasome homeostasis.

13.
ACS Infect Dis ; 2(11): 807-815, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27739665

ABSTRACT

Although serine proteases are important mediators of Mycobacterium tuberculosis (Mtb) virulence, there are currently no tools to selectively block or visualize members of this family of enzymes. Selective reporter substrates or activity-based probes (ABPs) could provide a means to monitor infection and response to therapy using imaging methods. Here, we use a combination of substrate selectivity profiling and focused screening to identify optimized reporter substrates and ABPs for the Mtb "Hydrolase important for pathogenesis 1" (Hip1) serine protease. Hip1 is a cell-envelope-associated enzyme with minimal homology to host proteases, making it an ideal target for probe development. We identified substituted 7-amino-4-chloro-3-(2-bromoethoxy)isocoumarins as irreversible inhibitor scaffolds. Furthermore, we used specificity data to generate selective reporter substrates and to further optimize a selective chloroisocoumarin inhibitor. These new reagents are potentially useful in delineating the roles of Hip1 during pathogenesis or as diagnostic imaging tools for specifically monitoring Mtb infections.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Enzyme Inhibitors/chemistry , Mycobacterium tuberculosis/enzymology , Serine Proteases/metabolism , Tuberculosis/microbiology , Animals , Antitubercular Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Design , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Serine Proteases/chemistry , Serine Proteases/genetics , Substrate Specificity
14.
Antimicrob Agents Chemother ; 59(1): 654-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25313210

ABSTRACT

wALADin1 benzimidazoles are specific inhibitors of δ-aminolevulinic acid dehydratase from Wolbachia endobacteria of filarial nematodes. We report that wALADin1 and two derivatives killed blood stage Plasmodium falciparum in vitro (50% inhibitory concentrations, 39, 7.7, and 12.8 µM, respectively). One of these derivatives inhibited gliding motility of Plasmodium berghei ANKA infectious sporozoites with nanomolar affinity and blocked invasion into hepatocytes but did not affect intrahepatocytic replication. Hence, wALADin1 benzimidazoles are tools to study gliding motility and potential antiplasmodial drug candidates.


Subject(s)
Antimalarials/pharmacology , Benzimidazoles/pharmacology , Plasmodium falciparum/drug effects , Porphobilinogen Synthase/antagonists & inhibitors , Benzimidazoles/chemistry , Humans , Inhibitory Concentration 50 , Plasmodium berghei/drug effects , Plasmodium falciparum/physiology , Thiophenes/chemistry , Thiophenes/pharmacology , Toxoplasma/drug effects
15.
J Med Chem ; 57(6): 2498-510, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24568185

ABSTRACT

The heme biosynthesis enzyme porphobilinogen synthase (PBGS) is a potential drug target in several human pathogens. wALADin1 benzimidazoles have emerged as species-selective PBGS inhibitors against Wolbachia endobacteria of filarial worms. In the present study, we have systematically tested wALADins against PBGS orthologs from bacteria, protozoa, metazoa, and plants to elucidate the inhibitory spectrum. However, the effect of wALADin1 on different PBGS orthologs was not limited to inhibition: several orthologs were stimulated by wALADin1; others remained unaffected. We demonstrate that wALADins allosterically modulate the PBGS homooligomeric equilibrium with inhibition mediated by favoring low-activity oligomers, while 5-aminolevulinic acid, Mg(2+), or K(+) stabilized high-activity oligomers. Pseudomonas aeruginosa PBGS could be inhibited or stimulated by wALADin1 depending on these factors and pH. We have defined the wALADin chemotypes responsible for either inhibition or stimulation, facilitating the design of tailored PBGS modulators for potential application as antimicrobial agents, herbicides, or drugs for porphyric disorders.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Porphobilinogen Synthase/antagonists & inhibitors , Animals , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/pharmacology , Chlamydia/drug effects , Herbicides/chemical synthesis , Herbicides/pharmacology , Humans , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Molecular Weight , Pisum sativum , Plants , Porphyrias/drug therapy , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Rickettsia/drug effects , Stereoisomerism , Structure-Activity Relationship , Wolbachia/drug effects
16.
Bioorg Med Chem Lett ; 23(20): 5558-62, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24012123

ABSTRACT

Substituted benzimidazoles of the wALADin1-family have recently been identified as a new class of species-selective inhibitors of delta-aminolevulinic acid dehydratase (ALAD) from Wolbachia endobacteria of parasitic filarial worms. Due to its Wolbachia-dependent antifilarial activity, wALADin1 is a starting point for the development of new drugs against filarial nematodes. We now present several other chemotypes of ALAD inhibitors that have been identified based upon their molecular similarity to wALADin1. A tricyclic quinoline derivative (wALADin2) with a different inhibitory mechanism and improved inhibitory potency and selectivity may represent an improved drug lead candidate.


Subject(s)
Benzimidazoles/chemistry , Enzyme Inhibitors/chemistry , Filaricides/chemistry , Porphobilinogen Synthase/antagonists & inhibitors , Thiophenes/chemistry , Wolbachia/enzymology , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/metabolism , Brugia malayi/enzymology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Filaricides/chemical synthesis , Filaricides/metabolism , Kinetics , Porphobilinogen Synthase/metabolism , Protein Binding , Quinolines/chemistry , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/metabolism
17.
Chem Biol ; 20(2): 177-87, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23438747

ABSTRACT

Lymphatic filariasis and onchocerciasis are severe diseases caused by filarial worms and affect more than 150 million people worldwide. Endosymbiotic α-proteobacteria Wolbachia are essential for these parasites throughout their life cycle. Using a high-throughput chemical screen, we identified a benzimidazole compound, wALADin1, that selectively targets the δ-aminolevulinic acid dehydratase (ALAD) of Wolbachia (wALAD) and exhibits macrofilaricidal effects on Wolbachia-containing filarial worms in vitro. wALADin1 is a mixed competitive/noncompetitive inhibitor that interferes with the Mg(2+)-induced activation of wALAD. This mechanism inherently excludes activity against the Zn(2+)-dependent human ortholog and might be translatable to Mg(2+)-responsive orthologs of other bacterial or protozoan pathogens. The specificity profile of wALADin1 derivatives reveals chemical features responsible for inhibitory potency and species selectivity. Our findings validate wALADins as a basis for developing potent leads that meet current requirements for antifilarial drugs.


Subject(s)
Antiprotozoal Agents/pharmacology , Benzimidazoles/pharmacology , Filarioidea/drug effects , Heme/biosynthesis , Thiophenes/pharmacology , Wolbachia/metabolism , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/therapeutic use , Benzimidazoles/chemistry , Benzimidazoles/therapeutic use , Drug Design , Elephantiasis, Filarial/drug therapy , High-Throughput Screening Assays , Humans , Kinetics , Magnesium/chemistry , Magnesium/metabolism , Porphobilinogen Synthase/antagonists & inhibitors , Porphobilinogen Synthase/metabolism , Symbiosis , Thiophenes/chemistry , Thiophenes/therapeutic use , Wolbachia/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...