Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Proc Natl Acad Sci U S A ; 121(16): e2309211121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38593081

ABSTRACT

Vesicular release of neurotransmitters and hormones relies on the dynamic assembly of the exocytosis/trans-SNARE complex through sequential interactions of synaptobrevins, syntaxins, and SNAP-25. Despite SNARE-mediated release being fundamental for intercellular communication in all excitable tissues, the role of auxiliary proteins modulating the import of reserve vesicles to the active zone, and thus, scaling repetitive exocytosis remains less explored. Secretagogin is a Ca2+-sensor protein with SNAP-25 being its only known interacting partner. SNAP-25 anchors readily releasable vesicles within the active zone, thus being instrumental for 1st phase release. However, genetic deletion of secretagogin impedes 2nd phase release instead, calling for the existence of alternative protein-protein interactions. Here, we screened the secretagogin interactome in the brain and pancreas, and found syntaxin-4 grossly overrepresented. Ca2+-loaded secretagogin interacted with syntaxin-4 at nanomolar affinity and 1:1 stoichiometry. Crystal structures of the protein complexes revealed a hydrophobic groove in secretagogin for the binding of syntaxin-4. This groove was also used to bind SNAP-25. In mixtures of equimolar recombinant proteins, SNAP-25 was sequestered by secretagogin in competition with syntaxin-4. Kd differences suggested that secretagogin could shape unidirectional vesicle movement by sequential interactions, a hypothesis supported by in vitro biological data. This mechanism could facilitate the movement of transport vesicles toward release sites, particularly in the endocrine pancreas where secretagogin, SNAP-25, and syntaxin-4 coexist in both α- and ß-cells. Thus, secretagogin could modulate the pace and fidelity of vesicular hormone release by differential protein interactions.


Subject(s)
Membrane Fusion , Secretagogins , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Secretagogins/metabolism , Cell Membrane/metabolism , Synaptosomal-Associated Protein 25/metabolism , Exocytosis , Cell Communication , Syntaxin 1/metabolism , Protein Binding
2.
J Am Chem Soc ; 143(10): 3794-3807, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33496585

ABSTRACT

Structural information is crucial for understanding catalytic mechanisms and to guide enzyme engineering efforts of biocatalysts, such as terpene cyclases. However, low sequence similarity can impede homology modeling, and inherent protein instability presents challenges for structural studies. We hypothesized that X-ray crystallography of engineered thermostable ancestral enzymes can enable access to reliable homology models of extant biocatalysts. We have applied this concept in concert with molecular modeling and enzymatic assays to understand the structure activity relationship of spiroviolene synthase, a class I terpene cyclase, aiming to engineer its specificity. Engineering a surface patch in the reconstructed ancestor afforded a template structure for generation of a high-confidence homology model of the extant enzyme. On the basis of structural considerations, we designed and crystallized ancestral variants with single residue exchanges that exhibited tailored substrate specificity and preserved thermostability. We show how the two single amino acid alterations identified in the ancestral scaffold can be transferred to the extant enzyme, conferring a specificity switch that impacts the extant enzyme's specificity for formation of the diterpene spiroviolene over formation of sesquiterpenes hedycaryol and farnesol by up to 25-fold. This study emphasizes the value of ancestral sequence reconstruction combined with enzyme engineering as a versatile tool in chemical biology.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Protein Engineering , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/genetics , Biocatalysis , Crystallography, X-Ray , Cyclization , Diterpenes/chemistry , Diterpenes/metabolism , Mutagenesis, Site-Directed , Protein Conformation , Sesquiterpenes/chemistry , Sesquiterpenes/metabolism , Substrate Specificity
3.
EMBO J ; 36(14): 2107-2125, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28637794

ABSTRACT

Ca2+-sensor proteins are generally implicated in insulin release through SNARE interactions. Here, secretagogin, whose expression in human pancreatic islets correlates with their insulin content and the incidence of type 2 diabetes, is shown to orchestrate an unexpectedly distinct mechanism. Single-cell RNA-seq reveals retained expression of the TRP family members in ß-cells from diabetic donors. Amongst these, pharmacological probing identifies Ca2+-permeable transient receptor potential vanilloid type 1 channels (TRPV1) as potent inducers of secretagogin expression through recruitment of Sp1 transcription factors. Accordingly, agonist stimulation of TRPV1s fails to rescue insulin release from pancreatic islets of glucose intolerant secretagogin knock-out(-/-) mice. However, instead of merely impinging on the SNARE machinery, reduced insulin availability in secretagogin-/- mice is due to ß-cell loss, which is underpinned by the collapse of protein folding and deregulation of secretagogin-dependent USP9X deubiquitinase activity. Therefore, and considering the desensitization of TRPV1s in diabetic pancreata, a TRPV1-to-secretagogin regulatory axis seems critical to maintain the structural integrity and signal competence of ß-cells.


Subject(s)
Gene Expression Regulation , Insulin-Secreting Cells/physiology , Proteins/metabolism , Secretagogins/metabolism , TRPV Cation Channels/metabolism , Animals , Cell Survival , Gene Expression Profiling , Humans , Mice , Mice, Knockout , Secretagogins/deficiency , Single-Cell Analysis
4.
Bioorg Med Chem ; 30: 115898, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33388594

ABSTRACT

The spread of antibiotic resistance within the ESKAPE group of human pathogenic bacteria poses severe challenges in the treatment of infections and maintenance of safe hospital environments. This motivates efforts to validate novel target proteins within these species that could be pursued as potential targets for antibiotic development. Genetic data suggest that the enzyme FabG, which is part of the bacterial fatty acid biosynthetic system FAS-II, is essential in several ESKAPE pathogens. FabG catalyzes the NADPH dependent reduction of 3-keto-acyl-ACP during fatty acid elongation, thus enabling lipid supply for production and maintenance of the cell envelope. Here we report on small-molecule screening on the FabG enzymes from A. baumannii and S. typhimurium to identify a set of µM inhibitors, with the most potent representative (1) demonstrating activity against six FabG-orthologues. A co-crystal structure with FabG from A. baumannii (PDB:6T65) confirms inhibitor binding at an allosteric site located in the subunit interface, as previously demonstrated for other sub-µM inhibitors of FabG from P. aeruginosa. We show that inhibitor binding distorts the oligomerization interface in the FabG tetramer and displaces crucial residues involved in the interaction with the co-substrate NADPH. These observations suggest a conserved allosteric site across the FabG family, which can be potentially targeted for interference with fatty acid biosynthesis in clinically relevant ESKAPE pathogens.


Subject(s)
Acinetobacter baumannii/enzymology , Alcohol Oxidoreductases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pseudomonas aeruginosa/enzymology , Salmonella typhimurium/enzymology , Alcohol Oxidoreductases/metabolism , Binding Sites/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Structure-Activity Relationship
5.
EMBO J ; 34(1): 36-54, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25430741

ABSTRACT

A hierarchical hormonal cascade along the hypothalamic-pituitary-adrenal axis orchestrates bodily responses to stress. Although corticotropin-releasing hormone (CRH), produced by parvocellular neurons of the hypothalamic paraventricular nucleus (PVN) and released into the portal circulation at the median eminence, is known to prime downstream hormone release, the molecular mechanism regulating phasic CRH release remains poorly understood. Here, we find a cohort of parvocellular cells interspersed with magnocellular PVN neurons expressing secretagogin. Single-cell transcriptome analysis combined with protein interactome profiling identifies secretagogin neurons as a distinct CRH-releasing neuron population reliant on secretagogin's Ca(2+) sensor properties and protein interactions with the vesicular traffic and exocytosis release machineries to liberate this key hypothalamic releasing hormone. Pharmacological tools combined with RNA interference demonstrate that secretagogin's loss of function occludes adrenocorticotropic hormone release from the pituitary and lowers peripheral corticosterone levels in response to acute stress. Cumulatively, these data define a novel secretagogin neuronal locus and molecular axis underpinning stress responsiveness.


Subject(s)
Corticosterone/metabolism , Corticotropin-Releasing Hormone/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Secretagogins/metabolism , Stress, Physiological/physiology , Animals , Corticosterone/genetics , Corticotropin-Releasing Hormone/genetics , Male , Mice , Neurons/cytology , Paraventricular Hypothalamic Nucleus/cytology , Pituitary Gland/cytology , Pituitary Gland/metabolism , RNA Interference , Secretagogins/genetics , Transcriptome/physiology
6.
Proteins ; 86(9): 912-923, 2018 09.
Article in English | MEDLINE | ID: mdl-29722065

ABSTRACT

RipA plays a vital role during cell division of Mycobacterium tuberculosis by degrading the cell wall peptidoglycan at the septum, allowing daughter cell separation. The peptidoglycan degrading activity relies on the NlpC/P60 domain, and as it is potentially harmful when deregulated, spatial and temporal control is necessary in this process. The N-terminal domain of RipA has been proposed to play an inhibitory role blocking the C-terminal NlpC/P60 domain. Accessibility of the active site cysteine residue is however not limited by the presence of the N-terminal domain, but by the lid-module of the inter-domain linker, which is situated in the peptide binding groove of the crystal structures of the catalytic domain. The 2.2 Å resolution structure of the N-terminal domain, determined by Se-SAD phasing, reveals an all-α-fold with 2 long α-helices, and shows similarity to bacterial periplasmic protein domains with scaffold-building role. Size exclusion chromatography and SAXS experiments are consistent with dimer formation of this domain in solution. The SAXS data from the periplasmic two-domain RipA construct suggest a rigid baton-like structure of the N-terminal module, with the catalytic domain connected by a 24 residue long flexible linker. This flexible linker allows for a catalytic zone, which is part of the spatiotemporal control of peptidoglycan degradation.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/enzymology , Hydrolases/metabolism , Bacterial Proteins/chemistry , Biocatalysis , Catalytic Domain , Hydrolases/chemistry , Mycobacterium tuberculosis/metabolism , Peptidoglycan/metabolism , Protein Conformation , Protein Multimerization
7.
Bioorg Med Chem Lett ; 27(19): 4582-4587, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28882483

ABSTRACT

CysK1 and CysK2 are two members of the cysteine/S-sulfocysteine synthase family in Mycobacterium tuberculosis, responsible for the de novo biosynthesis of l-cysteine, which is subsequently used as a building block for mycothiol. This metabolite is the first line defense of this pathogen against reactive oxygen and nitrogen species released by host macrophages after phagocytosis. In a previous medicinal chemistry campaign we had developed urea-based inhibitors of the cysteine synthase CysM with bactericidal activity against dormant M. tuberculosis. In this study we extended these efforts by examination of the in vitro activities of a library consisting of 71 urea compounds against CysK1 and CysK2. Binding was established by fluorescence spectroscopy and inhibition by enzyme assays. Several of the compounds inhibited these two cysteine synthases, with the most potent inhibitor displaying an IC50 value of 2.5µM for CysK1 and 6.6µM for CysK2, respectively. Four of the identified molecules targeting CysK1 and CysK2 were also among the top ten inhibitors of CysM, suggesting that potent compounds could be developed with activity against all three enzymes.


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Urea/pharmacology , Cysteine Synthase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
8.
Biochim Biophys Acta ; 1854(9): 1175-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25484279

ABSTRACT

The alarming increase of drug resistance in Mycobacterium tuberculosis strains poses a severe threat to human health. Chemotherapy is particularly challenging because M. tuberculosis can persist in the lungs of infected individuals; estimates of the WHO indicate that about 1/3 of the world population is infected with latent tuberculosis providing a large reservoir for relapse and subsequent spread of the disease. Persistent M. tuberculosis shows considerable tolerance towards conventional antibiotics making treatment particularly difficult. In this phase the bacilli are exposed to oxygen and nitrogen radicals generated as part of the host response and redox-defense mechanisms are thus vital for the survival of the pathogen. Sulfur metabolism and de novo cysteine biosynthesis have been shown to be important for the redox homeostasis in persistent M. tuberculosis and these pathways could provide promising targets for novel antibiotics for the treatment of the latent form of the disease. Recent research has provided evidence for three de novo metabolic routes of cysteine biosynthesis in M. tuberculosis, each with a specific PLP dependent cysteine synthase with distinct substrate specificities. In this review we summarize our present understanding of these pathways, with a focus on the advances on functional and mechanistic characterization of mycobacterial PLP dependent cysteine synthases, their role in the various pathways to cysteine, and first attempts to develop specific inhibitors of mycobacterial cysteine biosynthesis. This article is part of a Special Issue entitled: Cofactor-dependent proteins: evolution, chemical diversity and bio-applications.


Subject(s)
Cysteine Synthase/chemistry , Mycobacterium tuberculosis/enzymology , Pyridoxal Phosphate/physiology , Anti-Bacterial Agents/pharmacology , Cysteine/biosynthesis , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/metabolism , Humans , Mycobacterium tuberculosis/drug effects
9.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 3): 721-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25760619

ABSTRACT

Locus PA4043 in the genome of Pseudomonas aeruginosa PAO1 has been annotated as coding for a farnesyl pyrophosphate synthase (FPPS). This open reading frame was cloned and expressed recombinantly in Escherichia coli. The dimeric enzyme shows farnesyl pyrophosphate synthase activity and is strongly inhibited by ibandronate and zoledronate, drugs that are presently in clinical use. The structures of the unliganded enzyme and complexes with the substrate geranyl diphosphate (GPP), the inhibitor ibandronate and two compounds obtained from a differential scanning fluorimetry-based screen of a fragment library were determined by X-ray crystallography to resolutions of better than 2.0 Å. The enzyme shows the typical α-helical fold of farnesyl pyrophosphate synthases. The substrate GPP binds in the S1 substrate site in an open conformation of the enzyme. In the enzyme-ibandronate complex three inhibitor molecules are bound in the active site of the enzyme. One inhibitor molecule occupies the allylic substrate site (S1) of each subunit, as observed in complexes of nitrogen-containing bisphosphonate inhibitors of farnesyl synthases from other species. Two (in subunit A) and one (in subunit B) additional ibandronate molecules are bound in the active site. The structures of the fragment complexes show two molecules bound in a hydrophobic pocket adjacent to the active site. This allosteric pocket, which has previously only been described for FPPS from eukaryotic organisms, is thus also present in enzymes from pathogenic prokaryotes and might be utilized for the design of inhibitors of bacterial FPPS with a different chemical scaffold to the highly charged bisphosphonates, which are less likely to pass bacterial membranes.


Subject(s)
Bacterial Proteins/chemistry , Diphosphonates/chemistry , Enzyme Inhibitors/chemistry , Geranyltranstransferase/chemistry , Pseudomonas aeruginosa/enzymology , Bacterial Proteins/antagonists & inhibitors , Crystallography, X-Ray , Geranyltranstransferase/antagonists & inhibitors , Ibandronic Acid , Protein Structure, Secondary , Protein Structure, Tertiary
10.
Cereb Cortex ; 24(12): 3277-88, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23897649

ABSTRACT

The distribution and (patho-)physiological role of neuropeptides in the adult and aging brain have been extensively studied. Galanin is an inhibitory neuropeptide that can coexist with γ-aminobutyric acid (GABA) in the adult forebrain. However, galanin's expression sites, mode of signaling, impact on neuronal morphology, and colocalization with amino acid neurotransmitters during brain development are less well understood. Here, we show that galaninergic innervation of cholinergic projection neurons, which preferentially express galanin receptor 2 (GalR2) in the neonatal mouse basal forebrain, develops by birth. Nerve growth factor (NGF), known to modulate cholinergic morphogenesis, increases GalR2 expression. GalR2 antagonism (M871) in neonates reduces the in vivo expression and axonal targeting of the vesicular acetylcholine transporter (VAChT), indispensable for cholinergic neurotransmission. During cholinergic neuritogenesis in vitro, GalR2 can recruit Rho-family GTPases to induce the extension of a VAChT-containing primary neurite, the prospective axon. In doing so, GalR2 signaling dose-dependently modulates directional filopodial growth and antagonizes NGF-induced growth cone differentiation. Galanin accumulates in GABA-containing nerve terminals in the neonatal basal forebrain, suggesting its contribution to activity-driven cholinergic development during the perinatal period. Overall, our data define the cellular specificity and molecular complexity of galanin action in the developing basal forebrain.


Subject(s)
Cholinergic Neurons/cytology , Galanin/metabolism , Gene Expression Regulation, Developmental/physiology , Presynaptic Terminals/metabolism , Prosencephalon , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Cholinergic Neurons/drug effects , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Galanin/pharmacology , Glutamate Decarboxylase/genetics , Mice , Mice, Transgenic , Nerve Growth Factor/pharmacology , Presynaptic Terminals/drug effects , Prosencephalon/cytology , Prosencephalon/embryology , Prosencephalon/growth & development , Rats , Rats, Wistar , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism
11.
Biochem J ; 457(1): 33-41, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24107184

ABSTRACT

Enzymes carrying NlpC/p60 domains, for instance RipA and RipB from Mycobacterium tuberculosis, are bacterial peptidoglycan hydrolases that cleave the peptide stems and contribute to cell wall remodelling during cell division. A member of this protein family, RipD (Rv1566c) from M. tuberculosis described in the present study, displays sequence alterations in the NlpC/p60 catalytic triad and carries a pentapeptide repeat at its C-terminus. Bioinformatics analysis revealed RipD-like proteins in eleven mycobacterial genomes, whereas similar pentapeptide repeats occur in cell-wall-localized bacterial proteins and in a mycobacteriophage. In contrast with previously known members of the NlpC/p60 family, RipD does not show peptidoglycan hydrolase activity, which is consistent with the sequence alterations at the catalytic site. A strong interaction of the catalytically inactive core domain with peptidoglycan is however retained, presenting the first example of the NlpC/p60 domains that evolved to a non-catalytic peptidoglycan-binding function. Full-length RipD carrying the C-terminal repeat shows, however, a decrease in binding affinity to peptidoglycan, suggesting that the C-terminal tail modulates the interaction with bacterial cell wall components. The pentapeptide repeat at the C-terminus does not adopt a defined secondary structure in solution which is in accordance with results from the 1.17 Å (1 Å=0.1 nm) crystal structure of the protein carrying two repeat units.


Subject(s)
Adaptation, Biological/physiology , Mycobacterium tuberculosis/enzymology , N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Peptidoglycan/metabolism , Protein Interaction Domains and Motifs/physiology , Amino Acid Sequence , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Catalysis , Cell Wall/metabolism , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , N-Acetylmuramoyl-L-alanine Amidase/genetics , Protein Binding/physiology , Sequence Homology, Amino Acid
12.
J Bacteriol ; 196(19): 3410-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25022854

ABSTRACT

Mycobacterium tuberculosis is dependent on cysteine biosynthesis, and reduced sulfur compounds such as mycothiol synthesized from cysteine serve in first-line defense mechanisms against oxidative stress imposed by macrophages. Two biosynthetic routes to l-cysteine, each with its own specific cysteine synthase (CysK1 and CysM), have been described in M. tuberculosis, but the function of a third putative sulfhydrylase in this pathogen, CysK2, has remained elusive. We present biochemical and biophysical evidence that CysK2 is an S-sulfocysteine synthase, utilizing O-phosphoserine (OPS) and thiosulfate as substrates. The enzyme uses a mechanism via a central aminoacrylate intermediate that is similar to that of other members of this pyridoxal phosphate-dependent enzyme family. The apparent second-order rate of the first half-reaction with OPS was determined as kmax/Ks = (3.97 × 10(3)) ± 619 M(-1) s(-1), which compares well to the OPS-specific mycobacterial cysteine synthase CysM with a kmax/Ks of (1.34 × 10(3)) ± 48.2. Notably, CysK2 does not utilize thiocarboxylated CysO as a sulfur donor but accepts thiosulfate and sulfide as donor substrates. The specificity constant kcat/Km for thiosulfate is 40-fold higher than for sulfide, suggesting an annotation as S-sulfocysteine synthase. Mycobacterial CysK2 thus provides a third metabolic route to cysteine, either directly using sulfide as donor or indirectly via S-sulfocysteine. Hypothetically, S-sulfocysteine could also act as a signaling molecule triggering additional responses in redox defense in the pathogen upon exposure to reactive oxygen species during dormancy.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Lyases/chemistry , Lyases/metabolism , Mycobacterium tuberculosis/enzymology , Serine/metabolism , Bacterial Proteins/genetics , Kinetics , Lyases/genetics , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Serine/analogs & derivatives , Substrate Specificity
13.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 3): 432-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23519418

ABSTRACT

The transpeptidase LtdMt2 catalyzes the formation of the (3-3) cross-links characteristic of the peptidoglycan layer in the Mycobacterium tuberculosis cell wall. Bioinformatics analysis suggests that the extramembrane part of the enzyme consists of three domains: two smaller domains (denoted as A and B domains) and a transpeptidase domain (the C domain) at the C-terminus. The crystal structures of two fragments comprising the AB domains and the BC domains have been determined. The structure of the BC module, which was determined to 1.86 Šresolution using Se-SAD phasing, consists of the B domain with an immunoglobulin-related fold and the catalytic domain belonging to the ErfK/YbiS/YbnG fold family. The structure of the AB-domain fragment, which was solved by molecular replacement to 1.45 Šresolution, reveals that despite a lack of overall sequence identity the A domain is structurally very similar to the B domain. Combining the structures of the two fragments provides a view of the complete three-domain extramembrane part of LdtMt2 and shows that the protein extends at least 80-100 Šfrom the plasma membrane into the peptidoglycan layer and thus defines the maximal distance at which cross-links are formed by this enzyme. The LdtMt-related transpeptidases contain one or two immunoglobulin domains, which suggests that these might serve as extender units to position the catalytic domain at an appropriate distance from the membrane in the peptidoglycan layer.


Subject(s)
Bacterial Proteins/chemistry , Cell Wall/enzymology , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/chemistry , Aminoacyltransferases/chemistry , Catalytic Domain , Crystallography, X-Ray , Glycolipids/chemistry , Glycopeptides/chemistry , Models, Molecular , Peptidyl Transferases/classification , Protein Structure, Tertiary
14.
Bioorg Med Chem Lett ; 23(5): 1182-6, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23391589

ABSTRACT

Cysteine biosynthesis in Mycobacterium tuberculosis (MTB) is crucial for this pathogen to combat oxidative stress and for long term survival in the host. Hence inhibition of this pathway is attractive for developing novel drugs against tuberculosis. In the present study, the crystal structure of the mycobacterial enzyme O-acetylserine sulfhydrylase CysK1 bound to an oligopeptide inhibitor was used as a framework for virtual screening of the BITS-Pilani in-house database to identify new scaffolds as CysK1 inhibitors. Thirty compounds were synthesized and evaluated in vitro for their ability to inhibit CysK1, activity against M. tuberculosis and cytotoxicity as steps towards the derivation of structure-activity relationships (SAR) and lead optimization. Compound 8-nitro-4-(2-(trifluoromethyl)phenyl)-4,4a-dihydro-2H-pyrimido[5,4-e]thiazolo[3,2-a]pyrimidine-2,5(3H)-dione (4n) emerged as the most promising lead with an IC(50) of 17.7 µM for purified CysK1 and MIC of 7.6 µM for M. tuberculosis, with little or no cytotoxicity (>50 µM).


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Drug Design , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays/methods , Humans , Microbial Sensitivity Tests , Models, Molecular , Structure-Activity Relationship
15.
Article in English | MEDLINE | ID: mdl-23295481

ABSTRACT

Bacterial infections are increasingly difficult to treat owing to the spread of antibiotic resistance. A major concern is Gram-negative bacteria, for which the discovery of new antimicrobial drugs has been particularly scarce. In an effort to accelerate early steps in drug discovery, the EU-funded AEROPATH project aims to identify novel targets in the opportunistic pathogen Pseudomonas aeruginosa by applying a multidisciplinary approach encompassing target validation, structural characterization, assay development and hit identification from small-molecule libraries. Here, the strategies used for target selection are described and progress in protein production and structure analysis is reported. Of the 102 selected targets, 84 could be produced in soluble form and the de novo structures of 39 proteins have been determined. The crystal structures of eight of these targets, ranging from hypothetical unknown proteins to metabolic enzymes from different functional classes (PA1645, PA1648, PA2169, PA3770, PA4098, PA4485, PA4992 and PA5259), are reported here. The structural information is expected to provide a firm basis for the improvement of hit compounds identified from fragment-based and high-throughput screening campaigns.


Subject(s)
Bacterial Proteins/chemistry , Pseudomonas aeruginosa/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Catalytic Domain , Crystallography, X-Ray , Drug Discovery , Escherichia coli/genetics , Models, Molecular , Molecular Sequence Data , Protein Conformation
16.
ACS Chem Biol ; 18(4): 794-802, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37005433

ABSTRACT

Pseudouridimycin is a microbial C-nucleoside natural product that specifically inhibits bacterial RNA polymerases by binding to the active site and competing with uridine triphosphate for the nucleoside triphosphate (NTP) addition site. Pseudouridimycin consists of 5'-aminopseudouridine and formamidinylated, N-hydroxylated Gly-Gln dipeptide moieties to allow Watson-Crick base pairing and to mimic protein-ligand interactions of the triphosphates of NTP, respectively. The metabolic pathway of pseudouridimycin has been studied in Streptomyces species, but no biosynthetic steps have been characterized biochemically. Here, we show that the flavin-dependent oxidase SapB functions as a gate-keeper enzyme selecting pseudouridine (KM = 34 µM) over uridine (KM = 901 µM) in the formation of pseudouridine aldehyde. The pyridoxal phosphate (PLP)-dependent SapH catalyzes transamination, resulting in 5'-aminopseudouridine with a preference for arginine, methionine, or phenylalanine as cosubstrates as amino group donors. The binary structure of SapH in complex with pyridoxamine-5'-phosphate and site-directed mutagenesis identified Lys289 and Trp32 as key residues for catalysis and substrate binding, respectively. The related C-nucleoside oxazinomycin was accepted as a substrate by SapB with moderate affinity (KM = 181 µM) and was further converted by SapH, which opens possibilities for metabolic engineering to generate hybrid C-nucleoside pseudouridimycin analogues in Streptomyces.


Subject(s)
Nucleosides , Pseudouridine , Biosynthetic Pathways , DNA-Directed RNA Polymerases/metabolism , Nucleosides/metabolism , Pseudouridine/biosynthesis , Pseudouridine/metabolism , Pyridoxal Phosphate/chemistry , Streptomyces/chemistry , Streptomyces/metabolism
17.
Proteins ; 80(12): 2799-803, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22965870

ABSTRACT

NirD is part of the nitrite reductase complex NirBD that catalyses the reduction of nitrite to NH(3) in nitrate assimilation and anaerobic respiration. The crystal structure analysis of NirD from Mycobacterium tuberculosis shows a double ß-sandwich fold. NirD is related in three-dimensional structure and sequence to the Rieske proteins; however, it does not contain any Fe-S cluster or other cofactors that might be involved in electron transfer. A cysteine residue at the protein surface, conserved in NirD homologues lacking the iron-sulfur cluster might be important for the interaction with NirB and possibly stabilize one of the Fe-S centers in this subunit.


Subject(s)
Bacterial Proteins/chemistry , Mycobacterium tuberculosis/enzymology , Nitrite Reductases/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Crystallography, X-Ray , Molecular Sequence Data , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Nitrite Reductases/genetics , Protein Subunits , Sequence Alignment
18.
Eur J Med Chem ; 228: 113976, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34815129

ABSTRACT

Our previous studies on FabG have identified two compounds 5-bromo-2-(thiophene-2-carboxamido) benzoic acid (A) and ethyl 6-bromo-2-((dimethylamino)methyl)-5-hydroxy-1-phenyl-1H-indole-3-carboxylate(B) as best hits with allosteric mode of inhibition. FabG is an integral part of bacterial fatty acid biosynthetic system FAS II shown to be an essential gene in most ESKAPE Pathogens. The current work is focussed on lead expansion of these two hit molecules which ended up with forty-three analogues (twenty-nine analogues from lead compound A and fourteen compounds from lead compound B). The enzyme inhibition studies revealed that compound 15 (effective against EcFabG, AbFabG, StFabG, MtFabG1) and 19 (inhibiting EcFabG and StFabG) had potency of broad-spectrum inhibition on FabG panel.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Oxidoreductases/antagonists & inhibitors , Pseudomonas/drug effects , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Microbial Sensitivity Tests , Molecular Structure , Oxidoreductases/metabolism , Pseudomonas/enzymology , Structure-Activity Relationship
19.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 10): 1184-8, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-22102023

ABSTRACT

The ubiX gene (PA4019) of Pseudomonas aeruginosa has been annotated as encoding a putative 3-octaprenyl-4-hydroxybenzoate decarboxylase from the ubiquinone-biosynthesis pathway. Based on a transposon mutagenesis screen, this gene was also implicated as being essential for the survival of this organism. The crystal structure of recombinant UbiX determined to 1.5 Å resolution showed that the protein belongs to the superfamily of homo-oligomeric flavine-containing cysteine decarboxylases. The enzyme assembles into a dodecamer with 23 point symmetry. The subunit displays a typical Rossmann fold and contains one FMN molecule bound at the interface between two subunits.


Subject(s)
Carboxy-Lyases/chemistry , Pseudomonas aeruginosa/enzymology , Carboxy-Lyases/metabolism , Crystallography, X-Ray , Flavin Mononucleotide/chemistry , Flavin Mononucleotide/metabolism , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Subunits/chemistry , Structural Homology, Protein
20.
Cell Chem Biol ; 28(9): 1321-1332.e5, 2021 09 16.
Article in English | MEDLINE | ID: mdl-33826941

ABSTRACT

Effective treatment of tuberculosis is frequently hindered by the emerging antimicrobial resistance of Mycobacterium tuberculosis. The present study evaluates monocyclic ß-lactam compounds targeting the mycobacterial cell wall remodeling. Novel N-thio-ß-lactams were designed, synthesized, and characterized on the L,D-transpeptidase-2, a validated target in M. tuberculosis. The candidates were evaluated in biochemical assays identifying five compounds presenting target-specific kinetic constants equal or superior to meropenem, a carbapenem currently considered for tuberculosis therapy. Mass spectrometry in line with the crystal structures of five target-ligand complexes revealed that the N-thio-ß-lactams act via an unconventional mode of adduct formation, transferring the thio-residues from the lactam ring to the active-site cysteine of LdtMt2. The resulting stable adducts lead to a long-term inactivation of the target protein. Finally, the candidates were evaluated in vitro against a drug-susceptible and multidrug-resistant clinical isolates of M. tuberculosis, confirming the antimycobacterial effect of these novel compounds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Mycobacterium tuberculosis/drug effects , Peptidyl Transferases/antagonists & inhibitors , beta-Lactams/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/metabolism , beta-Lactams/chemical synthesis , beta-Lactams/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL