Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 92: 129387, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37369333

ABSTRACT

Metallo-ß-lactamases (MBLs) are a group of Zn(II)-dependent enzymes that pose a major threat to global health. They are linked to an increasing number of multi-drug resistant bacterial pathogens, but no clinically useful inhibitor is yet available. Since ß-lactam antibiotics, which are inactivated by MBLs, constitute ∼65% of all antibiotics used to treat infections, the search for clinically relevant MBL inhibitors is urgent. Here, derivatives of a 2-amino-1-benzyl-4,5-diphenyl-1H-pyrrole-3-carbonitrile (1a) were synthesised and their inhibitory effects assessed against prominent representatives of the MBL family. Several compounds are potent inhibitors of each MBL tested, making them promising candidates for the development of broad-spectrum drug leads. In particular, compound 5f is highly potent across the MBL family, with Ki values in the low µM range. Furthermore, this compound also appears to display synergy in combination with antibiotics such as penicillin G, cefuroxime or meropenem. This molecule thus represents a promising starting point to develop new drugs to inhibit a major mechanism of antibiotic resistance.


Subject(s)
beta-Lactamase Inhibitors , beta-Lactamases , beta-Lactamase Inhibitors/pharmacology , Anti-Bacterial Agents/pharmacology , Meropenem , Drug Resistance, Multiple, Bacterial
2.
Eur J Med Chem ; 254: 115383, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37087894

ABSTRACT

Purple acid phosphatases (PAPs) are ubiquitous binuclear metallohydrolases that have been isolated from various animals, plants and some types of fungi. In humans and mice, elevated PAP activity in osteoclasts is associated with osteoporosis, making human PAP an attractive target for the development of anti-osteoporotic drugs. Based on previous studies focusing on phosphonate scaffolds, as well as a new crystal structure of a PAP in complex with a derivative of a previously synthesized α-aminonaphthylmethylphosphonic acid, phosphonates 24-40 were designed as new PAP inhibitor candidates. Subsequent docking studies predicted that all of these compounds are likely to interact strongly with the active site of human PAP and most are likely to interact strongly with the active site of pig PAP. The seventeen candidates were synthesized with good yields and nine of them (26-28, 30, 33-36 and 38) inhibit in the sub-micromolar to nanomolar range against pig PAP, with 28 and 35 being the most potent mammalian PAP inhibitors reported with Ki values of 168 nM and 186 nM, respectively. This study thus paves the way for the next stage of drug development for phosphonate inhibitors of PAPs as anti-osteoporotic agents.


Subject(s)
Organophosphonates , Osteoporosis , Humans , Swine , Animals , Mice , Glycoproteins/chemistry , Acid Phosphatase , Plants , Organophosphonates/pharmacology , Mammals
3.
ChemMedChem ; 16(21): 3342-3359, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34331400

ABSTRACT

Metallohydrolases form a large group of enzymes that have fundamental importance in a broad range of biological functions. Among them, the purple acid phosphatases (PAPs) have gained attention due to their crucial role in the acquisition and use of phosphate by plants and also as a promising target for novel treatments of bone-related disorders and cancer. To date, no crystal structure of a mammalian PAP with drug-like molecules bound near the active site is available. Herein, we used a fragment-based design approach using structures of a mammalian PAP in complex with the MaybridgeTM fragment CC063346, the amino acid L-glutamine and the buffer molecule HEPES, as well as various solvent molecules to guide the design of highly potent and efficient mammalian PAP inhibitors. These inhibitors have improved aqueous solubility when compared to the clinically most promising PAP inhibitors available to date. Furthermore, drug-like fragments bound in newly discovered binding sites mapped out additional scaffolds for further inhibitor discovery, as well as scaffolds for the design of inhibitors with novel modes of action.


Subject(s)
Acid Phosphatase/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/pharmacology , Glutamine/pharmacology , Acid Phosphatase/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glutamine/chemical synthesis , Glutamine/chemistry , Ligands , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Swine
4.
Chem Biol Drug Des ; 97(5): 1048-1058, 2021 05.
Article in English | MEDLINE | ID: mdl-33455074

ABSTRACT

Cholinesterase inhibitors remain the mainstay of Alzheimer's disease treatment, and the search for new inhibitors with better efficacy and side effect profiles is ongoing. Virtual screening (VS) is a powerful technique for searching large compound databases for potential hits. This study used a sequential VS workflow combining ligand-based VS, molecular docking and physicochemical filtering to screen for central nervous system (CNS) drug-like acetylcholinesterase inhibitors (AChEIs) amongst the 6.9 million compounds of the CoCoCo database. Eleven in silico hits were initially selected, resulting in the discovery of an AChEI with a Ki of 3.2 µM. In vitro kinetics and in silico molecular dynamics experiments informed the selection of an additional seven analogues. This led to the discovery of two further AChEIs, with Ki values of 2.9 µM and 0.65 µM. All three compounds exhibited reversible, mixed inhibition of acetylcholinesterase. Importantly, the in silico physicochemical filter facilitated the discovery of CNS drug-like compounds, such that all three inhibitors displayed high in vitro blood-brain barrier model permeability.


Subject(s)
Acetylcholinesterase/chemistry , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Binding Sites , Butyrylcholinesterase/metabolism , Catalytic Domain , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Databases, Chemical , Donepezil/chemistry , Donepezil/metabolism , Donepezil/therapeutic use , Electrophorus/metabolism , Horses/metabolism , Kinetics , Molecular Dynamics Simulation , Permeability/drug effects
5.
J Med Chem ; 64(3): 1670-1684, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33512163

ABSTRACT

New drugs to treat tuberculosis (TB) are urgently needed to combat the increase in resistance observed among the current first-line and second-line treatments. Here, we propose ketol-acid reductoisomerase (KARI) as a target for anti-TB drug discovery. Twenty-two analogues of IpOHA, an inhibitor of plant KARI, were evaluated as antimycobacterial agents. The strongest inhibitor of Mycobacterium tuberculosis (Mt) KARI has a Ki value of 19.7 nM, fivefold more potent than IpOHA (Ki = 97.7 nM). This and four other potent analogues are slow- and tight-binding inhibitors of MtKARI. Three compounds were cocrystallized with Staphylococcus aureus KARI and yielded crystals that diffracted to 1.6-2.0 Å resolution. Prodrugs of these compounds possess antimycobacterial activity against H37Rv, a virulent strain of human TB, with the most active compound having an MIC90 of 2.32 ± 0.04 µM. This compound demonstrates a very favorable selectivity window and represents a highly promising lead as an anti-TB agent.


Subject(s)
Antitubercular Agents/pharmacology , Herbicides/pharmacology , Ketol-Acid Reductoisomerase/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Animals , Antitubercular Agents/chemistry , Cell Line , Cell Survival/drug effects , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Docking Simulation , Prodrugs , Staphylococcus aureus/enzymology
6.
Chemistry ; 27(9): 3130-3141, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33215746

ABSTRACT

New drugs aimed at novel targets are urgently needed to combat the increasing rate of drug-resistant tuberculosis (TB). Herein, the National Cancer Institute Developmental Therapeutic Program (NCI-DTP) chemical library was screened against a promising new target, ketol-acid reductoisomerase (KARI), the second enzyme in the branched-chain amino acid (BCAA) biosynthesis pathway. From this library, 6-hydroxy-2-methylthiazolo[4,5-d]pyrimidine-5,7(4H,6H)-dione (NSC116565) was identified as a potent time-dependent inhibitor of Mycobacterium tuberculosis (Mt) KARI with a Ki of 95.4 nm. Isothermal titration calorimetry studies showed that this inhibitor bound to MtKARI in the presence and absence of the cofactor, nicotinamide adenine dinucleotide phosphate (NADPH), which was confirmed by crystal structures of the compound in complex with closely related Staphylococcus aureus KARI. It is also shown that NSC116565 inhibits the growth of H37Ra and H37Rv strains of Mt with MIC50 values of 2.93 and 6.06 µm, respectively. These results further validate KARI as a TB drug target and show that NSC116565 is a promising lead for anti-TB drug development.


Subject(s)
Antitubercular Agents/pharmacology , Ketol-Acid Reductoisomerase/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Pyrimidinones/pharmacology , Cell Line , Humans , Ketol-Acid Reductoisomerase/metabolism , Mycobacterium tuberculosis/drug effects , NADP/metabolism , Staphylococcus aureus/enzymology , Tuberculosis/drug therapy , Tuberculosis/microbiology
7.
Bioorg Chem ; 105: 104386, 2020 12.
Article in English | MEDLINE | ID: mdl-33137556

ABSTRACT

Based on a structure-guided approach, aryl sulfonyl hydrazones conjugated with 1,3-diaryl pyrazoles were designed to target metallo-ß-lactamases (MBLs), using Klebsiella pneumoniaeNDM-1 as a model. The in vitro MBLs inhibition showed remarkable inhibition constant for most of the designed compounds at a low micromolar range (1.5-16.4 µM) against NDM-1, IMP-1 and AIM-1 MBLs. Furthermore, all compounds showed promising antibacterial activity against (K+, K1-K9) resistant clinical isolates of K. pneumoniae and were able to re-sensitize resistant K. pneumoniae (K5) strain towards meropenem and cefalexin. Besides, in vivo toxicity testing exhibited that the most active compound was non-toxic and well tolerated by the experimental animals orally up to 350 mg/kg and up to 125 mg/kg parenterally. The docking experiments on NDM-1 and IMP-1 rationalized the observed in vitro MBLs inhibition activity. Generally, this work presents a fruitful matrix to extend the chemical space for MBLs inhibition. This aids in tackling drug-resistance issues in antibacterial treatment.


Subject(s)
Anti-Bacterial Agents/pharmacology , Hydrazones/pharmacology , Klebsiella pneumoniae/drug effects , Pyrazoles/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Drug Design , Drug Resistance, Bacterial/drug effects , Hydrazones/chemical synthesis , Hydrazones/chemistry , Klebsiella pneumoniae/enzymology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Pyrazoles/chemistry , Structure-Activity Relationship , beta-Lactamase Inhibitors/chemical synthesis , beta-Lactamase Inhibitors/chemistry
8.
Bioorg Med Chem Lett ; 30(24): 127609, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33039562

ABSTRACT

Acetylcholinesterase inhibitors are the mainstay of Alzheimer's disease treatments, despite having only short-term symptomatic benefits and severe side effects. Selective butyrylcholinesterase inhibitors (BuChEIs) may be more effective treatments in late-stage Alzheimer's disease with fewer side effects. Virtual screening is a powerful tool for identifying potential inhibitors in large digital compound databases. This study used structure-based virtual screening combined with physicochemical filtering to screen the InterBioScreen and Maybridge databases for novel selective BuChEIs. The workflow rapidly identified 22 potential hits in silico, resulting in the discovery of a human BuChEI with low-micromolar potency in vitro (IC50 2.4 µM) and high selectivity for butyrylcholinesterase over acetylcholinesterase. The compound was a rapidly reversible BuChEI with mixed-model in vitro inhibition kinetics. The binding interactions were investigated using in silico molecular dynamics and by developing structure-activity relationships using nine analogues. The compound also displayed high permeability in an in vitro model of the blood-brain barrier.


Subject(s)
Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Drug Discovery , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Drug Design , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Structure-Activity Relationship
9.
Arch Biochem Biophys ; 692: 108516, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32745463

ABSTRACT

Ketol-acid reductoisomerase (KARI), the second enzyme in the branched-chain amino acid (BCAA) biosynthesis pathway, is an emerging target for the discovery of biocides. Here, we demonstrate that cyclopropane-1,1-dicarboxylate (CPD) inhibits KARIs from the pathogens Mycobacterium tuberculosis (Mt) and Campylobacter jejuni (Cj) reversibly with Ki values of 3.03 µM and 0.59 µM, respectively. Another reversible inhibitor of both KARIs, Hoe 704, is more potent than CPD with Ki values of 300 nM and 110 nM for MtKARI and CjKARI, respectively. The most potent inhibitor tested here is N-hydroxy-N-isopropyloxamate (IpOHA). It has a Ki of ~26 nM for MtKARI, but binds rather slowly (kon ~900 M-1s-1). In contrast, IpOHA binds more rapidly (kon ~7000 M-1s-1) to CjKARI and irreversibly.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Campylobacter jejuni/enzymology , Enzyme Inhibitors/chemistry , Ketol-Acid Reductoisomerase/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Campylobacter jejuni/chemistry , Cyclopropanes/chemistry , Dicarboxylic Acids/chemistry , Hydroxamic Acids/chemistry , Ketol-Acid Reductoisomerase/chemistry , Ketol-Acid Reductoisomerase/metabolism , Mycobacterium tuberculosis/chemistry , Organophosphorus Compounds/chemistry
10.
Nature ; 586(7828): 317-321, 2020 10.
Article in English | MEDLINE | ID: mdl-32640464

ABSTRACT

Acetohydroxyacid synthase (AHAS), also known as acetolactate synthase, is a flavin adenine dinucleotide-, thiamine diphosphate- and magnesium-dependent enzyme that catalyses the first step in the biosynthesis of branched-chain amino acids1. It is the target for more than 50 commercial herbicides2. AHAS requires both catalytic and regulatory subunits for maximal activity and functionality. Here we describe structures of the hexadecameric AHAS complexes of Saccharomyces cerevisiae and dodecameric AHAS complexes of Arabidopsis thaliana. We found that the regulatory subunits of these AHAS complexes form a core to which the catalytic subunit dimers are attached, adopting the shape of a Maltese cross. The structures show how the catalytic and regulatory subunits communicate with each other to provide a pathway for activation and for feedback inhibition by branched-chain amino acids. We also show that the AHAS complex of Mycobacterium tuberculosis adopts a similar structure, thus demonstrating that the overall AHAS architecture is conserved across kingdoms.


Subject(s)
Acetolactate Synthase/chemistry , Arabidopsis/enzymology , Saccharomyces cerevisiae/enzymology , Acetolactate Synthase/metabolism , Adenosine Triphosphate/metabolism , Amino Acids, Branched-Chain/biosynthesis , Catalytic Domain , Enzyme Activation , Evolution, Molecular , Feedback, Physiological , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mycobacterium tuberculosis/enzymology , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , Valine/metabolism
12.
Plant Sci ; 294: 110445, 2020 May.
Article in English | MEDLINE | ID: mdl-32234228

ABSTRACT

Phosphate acquisition by plants is an essential process that is directly implicated in the optimization of crop yields. Purple acid phosphatases (PAPs) are ubiquitous metalloenzymes, which catalyze the hydrolysis of a wide range of phosphate esters and anhydrides. While some plant PAPs display a preference for ATP as the substrate, others are efficient in hydrolyzing phytate or 2-phosphoenolpyruvate (PEP). PAP from red kidney bean (rkbPAP) is an efficient ATP- and ADPase, but has no activity towards phytate. Crystal structures of this enzyme in complex with ATP analogues (to 2.20 and 2.60 Å resolution, respectively) complement the recent structure of rkbPAP with a bound ADP analogue (ChemBioChem 20 (2019) 1536). Together these complexes provide the first structural insight of a PAP in complex with molecules that mimic biologically relevant substrates. Homology modeling was used to generate three-dimensional structures for the active sites of PAPs from tobacco (NtPAP) and thale cress (AtPAP26) that are efficient in hydrolyzing phytate and PEP as preferred substrates, respectively. The combining of crystallographic data, substrate docking simulations and a phylogenetic analysis of 49 plant PAP sequences (including the first PAP sequences reported from Eucalyptus) resulted in the identification of several active site residues that are important in defining the substrate specificities of plant PAPs; of particular relevance is the identification of a motif ("REKA") that is characteristic for plant PAPs that possess phytase activity. These results may inform bioengineering studies aimed at identifying and incorporating suitable plant PAP genes into crops to improve phosphorus acquisition and use efficiency. Organic phosphorus sources increasingly supplement or replace inorganic fertilizer, and efficient phosphorus use of crops will lower the environmental footprint of agriculture while enhancing food production.


Subject(s)
Acid Phosphatase/metabolism , Acid Phosphatase/genetics , Bioengineering/methods , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , Phaseolus/genetics , Phaseolus/metabolism , Substrate Specificity
13.
Chemistry ; 26(41): 8958-8968, 2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32198779

ABSTRACT

Ketol-acid reductoisomerase (KARI), the second enzyme in the branched-chain amino acid biosynthesis pathway, is a potential drug target for bacterial infections including Mycobacterium tuberculosis. Here, we have screened the Medicines for Malaria Venture Pathogen Box against purified M. tuberculosis (Mt) KARI and identified two compounds that have Ki values below 200 nm. In Mt cell susceptibility assays one of these compounds exhibited an IC50 value of 0.8 µm. Co-crystallization of this compound, 3-((methylsulfonyl)methyl)-2H-benzo[b][1,4]oxazin-2-one (MMV553002), in complex with Staphylococcus aureus KARI, which has 56 % identity with Mt KARI, NADPH and Mg2+ yielded a structure to 1.72 Šresolution. However, only a hydrolyzed product of the inhibitor (i.e. 3-(methylsulfonyl)-2-oxopropanic acid, missing the 2-aminophenol attachment) is observed in the active site. Surprisingly, Mt cell susceptibility assays showed that the 2-aminophenol product is largely responsible for the anti-TB activity of the parent compound. Thus, 3-(methylsulfonyl)-2-oxopropanic acid was identified as a potent KARI inhibitor that could be further explored as a potential biocidal agent and we have shown 2-aminophenol, as an anti-TB drug lead, especially given it has low toxicity against human cells. The study highlights that careful analysis of broad screening assays is required to correctly interpret cell-based activity data.


Subject(s)
Ketol-Acid Reductoisomerase/metabolism , Magnesium/chemistry , Mycobacterium tuberculosis/enzymology , NADP/chemistry , Staphylococcus aureus/metabolism , Catalytic Domain , Crystallization , Crystallography, X-Ray , Humans , Ketol-Acid Reductoisomerase/chemistry , Mycobacterium tuberculosis/chemistry , NADP/metabolism , Staphylococcus aureus/chemistry
14.
Eur J Med Chem ; 182: 111611, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31445230

ABSTRACT

Purple acid phosphatases (PAPs) are binuclear hydrolases that catalyze the hydrolysis of phosphorylated substrates under acidic to neutral conditions. Elevated serum concentrations of PAP are observed in patients suffering from osteoporosis, identifying this enzyme as a potential target for the development of novel therapeutic agents to treat this disease. α-Alkoxy-substituted naphthylmethylphosphonic acid derivatives have been identified previously as molecules that bind with high affinity to PAPs, and docking studies suggest that longer alkyl chains may increase the binding affinities of such compounds. Here, we synthesized several derivatives and tested their inhibitory effect against pig and red kidney bean PAPs. The most potent inhibitor within this series is the octadecyl derivative, which has a Ki value of ∼200 nM. Crystal structures of the dodecyl and octadecyl derivatives bound to red kidney bean PAP show that the length of the alkyl chain influences the ability of the phosphonate group to interact directly with the bimetallic center. These structures represent the first examples of potent inhibitors bound to a PAP that have drug-like properties. This study provides a starting point for the development of much needed new treatments for osteoporosis.


Subject(s)
Acid Phosphatase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Osteoporosis/drug therapy , Acid Phosphatase/metabolism , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Osteoporosis/metabolism , Phaseolus/enzymology , Structure-Activity Relationship , Swine
15.
Medchemcomm ; 10(1): 61-71, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30774855

ABSTRACT

Transgenic studies in animals have demonstrated a direct association between the level of expression of purple acid phosphatase (PAP; also known as tartrate-resistant acid phosphatase) and the progression of osteoporosis. Consequently, PAP has emerged as a promising target for the development of novel therapeutic agents to treat this debilitating disorder. PAPs are binuclear hydrolases that catalyse the hydrolysis of phosphorylated substrates under acidic to neutral conditions. A series of phenyltriazole carboxylic acids, prepared by the reactions of azide derivatives with propiolic acid through copper(i)-catalysed azide-alkyne cycloaddition click reactions, has been assessed for their inhibitory effect on the catalytic activity of pig and red kidney bean PAPs. The binding mode of most of these compounds is purely uncompetitive with K iuc values as low as ∼23 µM for the mammalian enzyme. Molecular modelling has been used to examine the binding modes of these triazole compounds in the presence of a substrate in the active site of the enzyme in order to rationalise their activities and to design more potent and specific derivatives.

16.
Chembiochem ; 20(12): 1536-1540, 2019 06 14.
Article in English | MEDLINE | ID: mdl-30719821

ABSTRACT

Purple acid phosphatases (PAPs) are members of the large family of metallohydrolases, a group of enzymes that perform a wide range of biological functions, while employing a highly conserved catalytic mechanism. PAPs are found in plants, animals and fungi; in humans they play an important role in bone turnover and are thus of interest for developing treatments for osteoporosis. The majority of metallohydrolases use a metal-bound hydroxide to initiate catalysis, which leads to the formation of a proposed five-coordinate oxyphosphorane species in the transition state. In this work, we crystallized PAP from red kidney beans (rkbPAP) in the presence of both adenosine and vanadate. The in crystallo-formed vanadate analogue of ADP provides detailed insight into the binding mode of a PAP substrate, captured in a structure that mimics the putative fivecoordinate transition state. Our observations not only provide unprecedented insight into the mechanism of metallohydrolases, but might also guide the structure-based design of inhibitors for application in the treatment of several human illnesses.


Subject(s)
Acid Phosphatase/chemistry , Plant Proteins/chemistry , Catalysis , Catalytic Domain , Kinetics , Models, Molecular , Phaseolus/metabolism , Vanadates/chemistry
17.
Eur J Med Chem ; 157: 462-479, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30107365

ABSTRACT

Purple acid phosphatases (PAPs) are metalloenzymes that catalyse the hydrolysis of phosphate esters under acidic conditions. Their active site contains a Fe(III)Fe(II) metal centre in mammals and a Fe(III)Zn(II) or Fe(III)Mn(II) metal centre in plants. In humans, elevated PAP levels in serum strongly correlate with the progression of osteoporosis and metabolic bone malignancies, which make PAP a target suitable for the development of chemotherapeutics to combat bone ailments. Due to difficulties in obtaining the human enzyme, the corresponding enzymes from red kidney bean and pig have been used previously to develop specific PAP inhibitors. Here, existing lead compounds were further elaborated to create a series of inhibitors with Ki values as low as ∼30 µM. The inhibition constants of these compounds were of comparable magnitude for pig and red kidney bean PAPs, indicating that relevant binding interactions are conserved. The crystal structure of red kidney bean PAP in complex with the most potent inhibitor in this series, compound 4f, was solved to 2.40 Šresolution. This inhibitor coordinates directly to the binuclear metal centre in the active site as expected based on its competitive mode of inhibition. Docking simulations predict that this compound binds to human PAP in a similar mode. This study presents the first example of a PAP structure in complex with an inhibitor that is of relevance to the development of anti-osteoporotic chemotherapeutics.


Subject(s)
Acid Phosphatase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Glycoproteins/antagonists & inhibitors , Osteoporosis/drug therapy , Osteoporosis/enzymology , Acid Phosphatase/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glycoproteins/metabolism , Molecular Docking Simulation , Molecular Structure , Phaseolus/enzymology , Structure-Activity Relationship , Swine
18.
Sci Rep ; 8(1): 3871, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29497067

ABSTRACT

Antibiotic resistance associated with the clinically significant carbapenemases KPC, NDM and OXA-48 in Enterobacteriaceae is emerging as worldwide. In Australia, IMP-producing Enterobacteriaceae are the most prevalent carbapenemase-producing Enterobacteriaceae (CPE). Genomic characteristics of such CPE are well described, but the corresponding proteome is poorly characterised. We have thus developed a method to analyse dynamic changes in the proteome of CPE under antibiotic pressure. Specifically, we have investigated the effect of meropenem at sub-lethal concentrations to develop a better understanding of how antibiotic pressure leads to resistance. Escherichia coli strains producing either NDM-, IMP- or KPC-type carbapenemases were included in this study, and their proteomes were analysed in growth conditions with or without meropenem. The most significant difference in the bacterial proteomes upon the addition of meropenem was triggered amongst NDM-producers and to a lower extent amongst KPC-producers. In particular, HU DNA-binding proteins, the GroEL/GroES chaperonin complex and GrpE proteins were overexpressed. These proteins may thus contribute to the better adaptability of NDM- and KPC-producers to meropenem. A significant meropenem-induced increase in the expression of the outer membrane protein A was only observed in IMP-producers, thus demonstrating that carbapenemase-mediated resistance relies on far more complex mechanisms than simple inactivation of the antibiotic.


Subject(s)
Escherichia coli/drug effects , Meropenem/pharmacology , Proteome/metabolism , Anti-Bacterial Agents/pharmacology , Australia , Bacteria/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbapenem-Resistant Enterobacteriaceae/drug effects , Enterobacteriaceae/drug effects , Enterobacteriaceae Infections/microbiology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Microbial Sensitivity Tests/methods , beta-Lactamases/genetics , beta-Lactamases/metabolism
19.
Chemistry ; 23(72): 18289-18295, 2017 Dec 22.
Article in English | MEDLINE | ID: mdl-28975665

ABSTRACT

Ketol-acid reductoisomerase (KARI) is an NAD(P)H and Mg2+ -dependent enzyme of the branched-chain amino acid (BCAA) biosynthesis pathway. Here, the first crystal structures of Staphylococcus aureus (Sa) KARI in complex with two transition state analogues, cyclopropane-1,1-dicarboxylate (CPD) and N-isopropyloxalyl hydroxamate (IpOHA) are reported. These compounds bind competitively and in multi-dentate manner to KARI with Ki values of 2.73 µm and 7.9 nm, respectively; however, IpOHA binds slowly to the enzyme. Interestingly, intact IpOHA is present in only ≈25 % of binding sites, whereas its deoxygenated form is present in the remaining sites. This deoxy form of IpOHA binds rapidly to Sa KARI, but with much weaker affinity (Ki =21 µm). Thus, our data pinpoint the origin of the slow binding mechanism of IpOHA. Furthermore, we propose that CPD mimics the early stage of the catalytic reaction (preceding the reduction step), whereas IpOHA mimics the late stage (after the reduction took place). These structural insights will guide strategies to design potent and rapidly binding derivatives of these compounds for the development of novel biocides.


Subject(s)
Bacterial Proteins/chemistry , Cyclopropanes/chemistry , Dicarboxylic Acids/chemistry , Hydroxamic Acids/chemistry , Ketol-Acid Reductoisomerase/chemistry , Staphylococcus aureus/enzymology , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Crystallization , Crystallography, X-Ray/methods , Ketol-Acid Reductoisomerase/metabolism , Models, Molecular , NAD/chemistry , Oxidation-Reduction , Protein Binding , Protein Conformation , Structure-Activity Relationship , Thermodynamics
20.
Org Biomol Chem ; 15(37): 7887-7893, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28891574

ABSTRACT

Recently we reported the aromatic Claisen rearrangements of benzyl ketene acetals, which form one of the few examples of aromatic Claisen rearrangements involving benzyl vinyl ethers (as opposed to allyl aryl ethers, which are the usual substrates for aromatic Claisen rearrangements). Theoretical calculations predict that these rearrangements proceed via a concerted [3,3]-sigmatropic transition state, which is similar in geometry to the TS for the Claisen rearrangement of an allyl aryl ether but has a 4 kcal mol-1 higher barrier. The effects of donor (OMe) and acceptor (CN) substituents on the kinetics of the [3,3]-rearrangement mirror those reported for allyl vinyl ethers: the largest substituent effects are seen for 1-OMe, 2-OMe, 2-CN, and 4-CN substituents, which lower the barrier by 5-9 kcal mol-1. Substituents on the aromatic ring have smaller effects on the barrier (≤2 kcal mol-1). The regioselectivities of Claisen rearrangements of meta-substituted benzyl ketene acetals favour 1,2,3-trisubstituted products in preference to the less sterically congested 1,2,4-isomers due to favourable orbital interactions in the 1,2,3 transition state.

SELECTION OF CITATIONS
SEARCH DETAIL
...