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










Publication year range
1.
J Biomol Struct Dyn ; 41(23): 14036-14048, 2023.
Article in English | MEDLINE | ID: mdl-36762717

ABSTRACT

Bipolar disorder is a major psychiatric disorder associated with cognitive impairment and a high suicide rate. Frontline therapy for this condition includes lithium (Li+)-containing treatments that can exert severe side effects. One target of Li+ is inositol monophosphatase-1 (IMPase1); inhibition of IMPase1 through small-molecule compounds may provide an alternative treatment for bipolar disorder. One such compound is the anti-inflammatory drug ebselen, which is well tolerated and safe; however, ebselen's exact mechanism of action in IMPase1 inhibition is not fully understood, preventing rational design of IMPase1 inhibitors. To fill this gap, we performed crystallographic and biochemical studies to investigate how ebselen inhibits IMPase1. We obtained a structure of IMPase1 in space group P21 after treatment with ebselen that revealed three key active-site loops (residues 33-44, 70-79, and 161-165) that are either disordered or in multiple conformations, supporting a hypothesis whereby dynamic conformational changes may be important for catalysis and ebselen inhibition. Using the thermal shift assay, we confirmed that ebselen significantly destabilizes the enzyme. Molecular docking suggests that ebselen could bind in the vicinity of His217. Investigation of the role of IMPase1 residues His217 and Cys218 suggests that inhibition of IMPase1 by ebselen may not be mediated via covalent modification of the active-site cysteine (Cys218) and is not affected by the covalent modification of other cysteine residues in the structure. Our results suggest that effects previously ascribed to ebselen-dependent inhibition likely result from disruption of essential active-site architecture, preventing activation of the IMPase1-Mg2+ complex.Communicated by Ramaswamy H. Sarma.


Subject(s)
Cysteine , Organoselenium Compounds , Humans , Molecular Docking Simulation , Phosphoric Monoester Hydrolases/chemistry , Lithium/pharmacology , Lithium/therapeutic use , Organoselenium Compounds/pharmacology , Organoselenium Compounds/chemistry
2.
FEBS J ; 290(9): 2412-2436, 2023 05.
Article in English | MEDLINE | ID: mdl-36178468

ABSTRACT

Fusarium endophytes damage cereal crops and contaminate produce with mycotoxins. Those fungi overcome the main chemical defence of host via detoxification by a malonyl-CoA-dependent enzyme homologous to xenobiotic metabolizing arylamine N-acetyltransferase (NAT). In Fusarium verticillioides (teleomorph Gibberella moniliformis, GIBMO), this N-malonyltransferase activity is attributed to (GIBMO)NAT1, and the fungus has two additional isoenzymes, (GIBMO)NAT3 (N-acetyltransferase) and (GIBMO)NAT2 (unknown function). We present the crystallographic structure of (GIBMO)NAT1, also modelling other fungal NAT homologues. Monomeric (GIBMO)NAT1 is distinctive, with access to the catalytic core through two "tunnel-like" entries separated by a "bridge-like" helix. In the quaternary arrangement, (GIBMO)NAT1 monomers interact in pairs along an extensive interface whereby one entry of each monomer is covered by the N-terminus of the other monomer. Although monomeric (GIBMO)NAT1 apparently accommodates acetyl-CoA better than malonyl-CoA, dimerization changes the active site to allow malonyl-CoA to reach the catalytic triad (Cys110, His158 and Asp173) via the single uncovered entry, and anchor its terminal carboxyl-group via hydrogen bonds to Arg109, Asn157 and Thr261. Lacking a terminal carboxyl-group, acetyl-CoA cannot form such stabilizing interactions, while longer acyl-CoAs enter the active site but cannot reach catalytic Cys. Other NAT isoenzymes lack such structural features, with (GIBMO)NAT3 resembling bacterial NATs and (GIBMO)NAT2 adopting a structure intermediate between (GIBMO)NAT1 and (GIBMO)NAT3. Biochemical assays confirmed differential donor substrate preference of (GIBMO)NAT isoenzymes, with phylogenetic analysis demonstrating evolutionary separation. Given the role of (GIBMO)NAT1 in enhancing Fusarium pathogenicity, unravelling the structure and function of this enzyme may benefit research into more targeted strategies for pathogen control.


Subject(s)
Arylamine N-Acetyltransferase , Fusarium , Arylamine N-Acetyltransferase/chemistry , Arylamine N-Acetyltransferase/genetics , Fusarium/genetics , Isoenzymes/genetics , Phylogeny , Acetyl Coenzyme A , Acetyltransferases
3.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 11): 707-713, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31702584

ABSTRACT

Domain swapping is a widespread oligomerization process that is observed in a large variety of protein families. In the large superfamily of substrate-binding proteins, non-monomeric members have rarely been reported. The arginine-binding protein from Thermotoga maritima (TmArgBP), a protein endowed with a number of unusual properties, presents a domain-swapped structure in its dimeric native state in which the two polypeptide chains mutually exchange their C-terminal helices. It has previously been shown that mutations in the region connecting the last two helices of the TmArgBP structure lead to the formation of a variety of oligomeric states (monomers, dimers, trimers and larger aggregates). With the aim of defining the structural determinants of domain swapping in TmArgBP, the monomeric form of the P235GK mutant has been structurally characterized. Analysis of this arginine-bound structure indicates that it consists of a closed monomer with its C-terminal helix folded against the rest of the protein, as typically observed for substrate-binding proteins. Notably, the two terminal helices are joined by a single nonhelical residue (Gly235). Collectively, the present findings indicate that extending the hinge region and conferring it with more conformational freedom makes the formation of a closed TmArgBP monomer possible. On the other hand, the short connection between the helices may explain the tendency of the protein to also adopt alternative oligomeric states (dimers, trimers and larger aggregates). The data reported here highlight the importance of evolutionary control to avoid the uncontrolled formation of heterogeneous and potentially harmful oligomeric species through domain swapping.


Subject(s)
Arginine/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Thermotoga maritima/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Crystallization , Mutation/genetics , Protein Binding , Structural Homology, Protein
4.
J Mol Recognit ; 30(11)2017 11.
Article in English | MEDLINE | ID: mdl-28608547

ABSTRACT

The Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging virus that poses a major challenge to clinical management. The 3C-like protease (3CLpro ) is essential for viral replication and thus represents a potential target for antiviral drug development. Presently, very few data are available on MERS-CoV 3CLpro inhibition by small molecules. We conducted extensive exploration of the pharmacophoric space of a recently identified set of peptidomimetic inhibitors of the bat HKU4-CoV 3CLpro . HKU4-CoV 3CLpro shares high sequence identity (81%) with the MERS-CoV enzyme and thus represents a potential surrogate model for anti-MERS drug discovery. We used 2 well-established methods: Quantitative structure-activity relationship (QSAR)-guided modeling and docking-based comparative intermolecular contacts analysis. The established pharmacophore models highlight structural features needed for ligand recognition and revealed important binding-pocket regions involved in 3CLpro -ligand interactions. The best models were used as 3D queries to screen the National Cancer Institute database for novel nonpeptidomimetic 3CLpro inhibitors. The identified hits were tested for HKU4-CoV and MERS-CoV 3CLpro inhibition. Two hits, which share the phenylsulfonamide fragment, showed moderate inhibitory activity against the MERS-CoV 3CLpro and represent a potential starting point for the development of novel anti-MERS agents. To the best of our knowledge, this is the first pharmacophore modeling study supported by in vitro validation on the MERS-CoV 3CLpro . HIGHLIGHTS: MERS-CoV is an emerging virus that is closely related to the bat HKU4-CoV. 3CLpro is a potential drug target for coronavirus infection. HKU4-CoV 3CLpro is a useful surrogate model for the identification of MERS-CoV 3CLpro enzyme inhibitors. dbCICA is a very robust modeling method for hit identification. The phenylsulfonamide scaffold represents a potential starting point for MERS coronavirus 3CLpro inhibitors development.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/enzymology , Chiroptera/virology , Middle East Respiratory Syndrome Coronavirus/drug effects , Protease Inhibitors/pharmacology , Viral Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Betacoronavirus/drug effects , Binding Sites , Computer Simulation , Ligands , Models, Molecular , Protease Inhibitors/chemistry , Quantitative Structure-Activity Relationship , ROC Curve , Reproducibility of Results , Viral Proteins/chemistry
5.
Br J Pharmacol ; 174(14): 2209-2224, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28380256

ABSTRACT

BACKGROUND AND PURPOSE: With the emergence of extensively drug-resistant tuberculosis, there is a need for new anti-tubercular drugs that work through novel mechanisms of action. The meta cleavage product hydrolase, HsaD, has been demonstrated to be critical for the survival of Mycobacterium tuberculosis in macrophages and is encoded in an operon involved in cholesterol catabolism, which is identical in M. tuberculosis and M. bovis BCG. EXPERIMENTAL APPROACH: We generated a mutant strain of M. bovis BCG with a deletion of hsaD and tested its growth on cholesterol. Using a fragment based approach, over 1000 compounds were screened by a combination of differential scanning fluorimetry, NMR spectroscopy and enzymatic assay with pure recombinant HsaD to identify potential inhibitors. We used enzymological and structural studies to investigate derivatives of the inhibitors identified and to test their effects on growth of M. bovis BCG and M. tuberculosis. KEY RESULTS: The hsaD deleted strain was unable to grow on cholesterol as sole carbon source but did grow on glucose. Of seven chemically distinct 'hits' from the library, two chemical classes of fragments were found to bind in the vicinity of the active site of HsaD by X-ray crystallography. The compounds also inhibited growth of M. tuberculosis on cholesterol. The most potent inhibitor of HsaD was also found to be the best inhibitor of mycobacterial growth on cholesterol-supplemented minimal medium. CONCLUSIONS AND IMPLICATIONS: We propose that HsaD is a novel therapeutic target, which should be fully exploited in order to design and discover new anti-tubercular drugs. LINKED ARTICLES: This article is part of a themed section on Drug Metabolism and Antibiotic Resistance in Micro-organisms. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.14/issuetoc.


Subject(s)
Antitubercular Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hydrolases/deficiency , Hydrolases/metabolism , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Structure-Activity Relationship
6.
Br J Pharmacol ; 174(14): 2194-2208, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28002883

ABSTRACT

Tuberculosis (TB), although a curable disease, is still one of the most difficult infections to treat. Mycobacterium tuberculosis infects 10 million people worldwide and kills 1.5 million people each year. Reactivation of a latent infection is the major cause of TB. Cholesterol is a critical carbon source during latent infection. Catabolism of cholesterol contributes to the pool of propionyl-CoA, a precursor that is incorporated into lipid virulence factors. The M. tuberculosis genome contains a large regulon of cholesterol catabolic genes suggesting that the microorganism can utilize host sterol for infection and persistence. The protein products of these genes present ideal targets for rational drug discovery programmes. This review summarizes the development of enzyme inhibitors targeting the cholesterol pathway in M. tuberculosis. This knowledge is essential for the discovery of novel agents to treat M. tuberculosis infection. LINKED ARTICLES: This article is part of a themed section on Drug Metabolism and Antibiotic Resistance in Micro-organisms. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.14/issuetoc.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cholesterol/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Tuberculosis/drug therapy , Tuberculosis/microbiology , Anti-Bacterial Agents/chemistry , Cholesterol/chemistry , Drug Discovery , Humans , Mycobacterium tuberculosis/genetics
7.
Future Med Chem ; 8(5): 509-26, 2016 04.
Article in English | MEDLINE | ID: mdl-27105126

ABSTRACT

Recent evidence points to significant roles played by protein kinases in cell signaling and cellular proliferation. Faulty protein kinases are involved in cancer, diabetes and chronic inflammation. Efforts are continuously carried out to discover new inhibitors for selected protein kinases. In this review, we discuss two new computer-aided methodologies we developed to mine virtual databases for new bioactive compounds. One method is ligand-based exploration of the pharmacophoric space of inhibitors of any particular biotarget followed by quantitative structure-activity relationship-based selection of the best pharmacophore(s). The second approach is structure-based assuming that potent ligands come into contact with binding site spots distinct from those contacted by weakly potent ligands. Both approaches yield pharmacophores useful as 3D search queries for the discovery of new bioactive (kinase) inhibitors.


Subject(s)
Drug Design , Drug Discovery/methods , Protein Kinase Inhibitors/chemistry , Protein Kinases/metabolism , Binding Sites , Computers , Ligands , Models, Molecular , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Quantitative Structure-Activity Relationship
8.
Expert Opin Drug Discov ; 11(2): 197-214, 2016.
Article in English | MEDLINE | ID: mdl-26558613

ABSTRACT

INTRODUCTION: Type-II diabetes mellitus (T2DM) is a complex chronic disease that represents a major therapeutic challenge. Despite extensive efforts in T2DM drug development, therapies remain unsatisfactory. Currently, there are many novel and important antidiabetic drug targets under investigation by many research groups worldwide. One of the main challenges to develop effective orally active hypoglycemic agents is off-target effects. Computational tools have impacted drug discovery at many levels. One of the earliest methods is quantitative structure-activity relationship (QSAR) studies. QSAR strategies help medicinal chemists understand the relationship between hypoglycemic activity and molecular properties. Hence, QSAR may hold promise in guiding the synthesis of specifically designed novel ligands that demonstrate high potency and target selectivity. AREAS COVERED: This review aims to provide an overview of the QSAR strategies used to model antidiabetic agents. In particular, this review focuses on drug targets that raised recent scientific interest and/or led to successful antidiabetic agents in the market. Special emphasis has been made on studies that led to the identification of novel antidiabetic scaffolds. EXPERT OPINION: Computer-aided molecular design and discovery techniques like QSAR have a great potential in designing leads against complex diseases such as T2DM. Combined with other in silico techniques, QSAR can provide more useful and rational insights to facilitate the discovery of novel compounds. However, since T2DM is a complex disease that includes several faulty biological targets, multi-target QSAR studies are recommended in the future to achieve efficient antidiabetic therapies.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Discovery/methods , Hypoglycemic Agents/therapeutic use , Animals , Computer Simulation , Computer-Aided Design , Diabetes Mellitus, Type 2/physiopathology , Drug Design , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Molecular Targeted Therapy , Quantitative Structure-Activity Relationship
9.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 11): 3066-79, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25372695

ABSTRACT

Mycobacterium abscessus is the most pathogenic rapid-growing mycobacterium and is one of the most resistant organisms to chemotherapeutic agents. However, structural and functional studies of M. abscessus proteins that could modify/inactivate antibiotics remain nonexistent. Here, the structural and functional characterization of an arylamine N-acetyltransferase (NAT) from M. abscessus [(MYCAB)NAT1] are reported. This novel prokaryotic NAT displays significant N-acetyltransferase activity towards aromatic substrates, including antibiotics such as isoniazid and p-aminosalicylate. The enzyme is endogenously expressed and functional in both the rough and smooth M. abscessus morphotypes. The crystal structure of (MYCAB)NAT1 at 1.8 Šresolution reveals that it is more closely related to Nocardia farcinica NAT than to mycobacterial isoforms. In particular, structural and physicochemical differences from other mycobacterial NATs were found in the active site. Peculiarities of (MYCAB)NAT1 were further supported by kinetic and docking studies showing that the enzyme was poorly inhibited by the piperidinol inhibitor of mycobacterial NATs. This study describes the first structure of an antibiotic-modifying enzyme from M. abscessus and provides bases to better understand the substrate/inhibitor-binding specificities among mycobacterial NATs and to identify/optimize specific inhibitors. These data should also contribute to the understanding of the mechanisms that are responsible for the pathogenicity and extensive chemotherapeutic resistance of M. abscessus.


Subject(s)
Arylamine N-Acetyltransferase/chemistry , Mycobacterium/enzymology , Acetylation , Amino Acid Sequence , Arylamine N-Acetyltransferase/genetics , Arylamine N-Acetyltransferase/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Mycobacterium/chemistry , Mycobacterium/genetics , Mycobacterium/metabolism , Mycobacterium Infections/microbiology , Phylogeny , Substrate Specificity
10.
Molecules ; 19(10): 16274-90, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25310152

ABSTRACT

Novel drugs to treat tuberculosis are required and the identification of potential targets is important. Piperidinols have been identified as potential antimycobacterial agents (MIC < 5 µg/mL), which also inhibit mycobacterial arylamine N-acetyltransferase (NAT), an enzyme essential for mycobacterial survival inside macrophages. The NAT inhibition involves a prodrug-like mechanism in which activation leads to the formation of bioactive phenyl vinyl ketone (PVK). The PVK fragment selectively forms an adduct with the cysteine residue in the active site. Time dependent inhibition of the NAT enzyme from Mycobacterium marinum (M. marinum) demonstrates a covalent binding mechanism for all inhibitory piperidinol analogues. The structure activity relationship highlights the importance of halide substitution on the piperidinol benzene ring. The structures of the NAT enzymes from M. marinum and M. tuberculosis, although 74% identical, have different residues in their active site clefts and allow the effects of amino acid substitutions to be assessed in understanding inhibitory potency. In addition, we have used the piperidinol 3-dimensional shape and electrostatic properties to identify two additional distinct chemical scaffolds as inhibitors of NAT. While one of the scaffolds has anti-tubercular activity, both inhibit NAT but through a non-covalent mechanism.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/metabolism , Binding Sites , Humans , Molecular Conformation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Protein Binding
11.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 8): 1433-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23897467

ABSTRACT

Arylamine N-acetyltransferase from Mycobacterium tuberculosis (TBNAT) plays an important role in the intracellular survival of the microorganism inside macrophages. Medicinal chemistry efforts to optimize inhibitors of the TBNAT enzyme have been hampered by the lack of a three-dimensional structure of the enzyme. In this paper, the first structure of TBNAT, determined using a lone crystal produced using cross-seeding with the homologous protein from M. marinum, is reported. Despite the similarity between the two enzymes (74% sequence identity), they show distinct physical and biochemical characteristics. The structure elegantly reveals the characteristic features of the protein surface as well as details of the active site of TBNAT relevant to drug-discovery efforts. The crystallographic analysis of the diffraction data presented many challenges, since the crystal was twinned and the habit possessed pseudo-translational symmetry.


Subject(s)
Arylamine N-Acetyltransferase/chemistry , Arylamine N-Acetyltransferase/metabolism , Mycobacterium marinum/enzymology , Mycobacterium tuberculosis/enzymology , Arylamine N-Acetyltransferase/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Crystallization/methods , Crystallography, X-Ray , Enzyme Stability , Models, Molecular , Protein Conformation , Scattering, Small Angle , Sequence Homology, Amino Acid
12.
PLoS One ; 7(12): e52790, 2012.
Article in English | MEDLINE | ID: mdl-23285185

ABSTRACT

Latent M. tuberculosis infection presents one of the major obstacles in the global eradication of tuberculosis (TB). Cholesterol plays a critical role in the persistence of M. tuberculosis within the macrophage during latent infection. Catabolism of cholesterol contributes to the pool of propionyl-CoA, a precursor that is incorporated into cell-wall lipids. Arylamine N-acetyltransferase (NAT) is encoded within a gene cluster that is involved in the cholesterol sterol-ring degradation and is essential for intracellular survival. The ability of the NAT from M. tuberculosis (TBNAT) to utilise propionyl-CoA links it to the cholesterol-catabolism pathway. Deleting the nat gene or inhibiting the NAT enzyme prevents intracellular survival and results in depletion of cell-wall lipids. TBNAT has been investigated as a potential target for TB therapies. From a previous high-throughput screen, 3-benzoyl-4-phenyl-1-methylpiperidinol was identified as a selective inhibitor of prokaryotic NAT that exhibited antimycobacterial activity. The compound resulted in time-dependent irreversible inhibition of the NAT activity when tested against NAT from M. marinum (MMNAT). To further evaluate the antimycobacterial activity and the NAT inhibition of this compound, four piperidinol analogues were tested. All five compounds exert potent antimycobacterial activity against M. tuberculosis with MIC values of 2.3-16.9 µM. Treatment of the MMNAT enzyme with this set of inhibitors resulted in an irreversible time-dependent inhibition of NAT activity. Here we investigate the mechanism of NAT inhibition by studying protein-ligand interactions using mass spectrometry in combination with enzyme analysis and structure determination. We propose a covalent mechanism of NAT inhibition that involves the formation of a reactive intermediate and selective cysteine residue modification. These piperidinols present a unique class of antimycobacterial compounds that have a novel mode of action different from known anti-tubercular drugs.


Subject(s)
Antitubercular Agents/pharmacology , Arylamine N-Acetyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Macrophages/microbiology , Mycobacterium/drug effects , Mycobacterium/enzymology , Piperidines/pharmacology , Animals , Antitubercular Agents/chemistry , Arylamine N-Acetyltransferase/chemistry , Arylamine N-Acetyltransferase/metabolism , Catalytic Domain , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Humans , Mice , Molecular Docking Simulation , Piperidines/chemistry , Protein Conformation
13.
Protein Expr Purif ; 80(2): 246-52, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21767648

ABSTRACT

Arylamine N-acetyltransferase from Mycobacterium tuberculosis (TBNAT) has been proposed as a drug target for latent tuberculosis treatment. The enzyme is essential for the survival of the mycobacterium in macrophages. However, TBNAT has been very difficult to generate as a soluble protein. In this work we describe production of soluble recombinant TBNAT at a reasonable yield achieved by subcloning the tbnat gene with a purification His-tag into the pVLT31 plasmid, and subsequent optimisation of the induction conditions. The expression system results in soluble protein optimised upon extended (60 h) low level isopropyl ß-D-1-thiogalactopyranoside level induction (100 µM) at a temperature of 15 °C. The level of TBNAT expression obtained in E. coli has been significantly improved from ∼2 mg to a final yield of up to 16 mg per litre of culture at a purity level suitable for structural studies. The molecular mass of 31310 Da was confirmed using mass spectroscopy and the oligomerisation state was determined. The stability of TBNAT in different buffer systems was investigated by thermal shift assays and sufficient protein is now available for the screening of chemical libraries for inhibitors.


Subject(s)
Arylamine N-Acetyltransferase/isolation & purification , Bacterial Proteins/isolation & purification , Mycobacterium tuberculosis/enzymology , Recombinant Proteins/isolation & purification , Arylamine N-Acetyltransferase/metabolism , Bacterial Proteins/metabolism , Buffers , Chromatography, Gel , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Enzyme Assays , Escherichia coli/genetics , Escherichia coli/metabolism , Histidine/metabolism , Isopropyl Thiogalactoside/pharmacology , Molecular Weight , Plasmids/genetics , Plasmids/metabolism , Protein Stability , Recombinant Proteins/metabolism , Solubility , Spectrometry, Mass, Electrospray Ionization , Temperature
14.
Bioorg Med Chem Lett ; 21(4): 1185-90, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21251821

ABSTRACT

The synthesis and inhibitory potencies of a novel series of ß-amino alcohols, based on the hit-compound 3-[3'-(4''-cyclopent-2'''-en-1'''-ylphenoxy)-2'-hydroxypropyl]-5,5 dimethylimidazolidine-2,4-dione as specific inhibitors of mycobacterial N-acetyltransferase (NAT) enzymes are reported. Effects of synthesised compounds on growth of Mycobacterium tuberculosis have been determined.


Subject(s)
Amino Alcohols/chemistry , Antitubercular Agents/chemistry , Arylamine N-Acetyltransferase/antagonists & inhibitors , Amino Alcohols/chemical synthesis , Amino Alcohols/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Arylamine N-Acetyltransferase/metabolism , Binding Sites , Catalytic Domain , Computer Simulation , Mycobacterium/drug effects , Mycobacterium/enzymology , Structure-Activity Relationship
15.
Article in English | MEDLINE | ID: mdl-20057057

ABSTRACT

Azoreductase 1 from Pseudomonas aeruginosa strain PAO1 (paAzoR1) catalyses the activation of the prodrug balsalazide and reduces the azo dye methyl red using reduced nicotinamide adenine dinucleotide cofactor as an electron donor. To investigate the mechanism of the enzyme, a Y131F mutation was introduced and the enzymic properties of the mutant were compared with those of the wild-type enzyme. The crystallographic structure of the mutant with methyl red bound was solved at 2.1 A resolution and compared with the wild-type structure. Tyr131 is important in the architecture of the active site but is not essential for enzymic activity.


Subject(s)
NADH, NADPH Oxidoreductases/chemistry , Tyrosine/chemistry , Azo Compounds , Catalysis , Catalytic Domain/genetics , Crystallography, X-Ray , Enzyme Stability , Hot Temperature , Kinetics , Mesalamine/metabolism , Mutagenesis, Site-Directed , NAD/metabolism , NADH, NADPH Oxidoreductases/metabolism , Nitroreductases , Phenylhydrazines/metabolism , Protein Folding , Pseudomonas aeruginosa/enzymology
16.
Protein Cell ; 1(4): 384-392, 2010 Apr.
Article in English | MEDLINE | ID: mdl-21203950

ABSTRACT

Treatment of latent tuberculosis infection remains an important goal of global TB eradication. To this end, targets that are essential for intracellular survival of Mycobacterium tuberculosis are particularly attractive. Arylamine N-acetyltransferase (NAT) represents such a target as it is, along with the enzymes encoded by the associated gene cluster, essential for mycobacterial survival inside macrophages and involved in cholesterol degradation. Cholesterol is likely to be the fuel for M. tuberculosis inside macrophages. Deleting the nat gene and inhibiting the NAT enzyme prevents survival of the microorganism in macrophages and induces cell wall alterations, rendering the mycobacterium sensitive to antibiotics to which it is normally resistant. To date, NAT from M. marinum (MMNAT) is considered the best available model for NAT from M. tuberculosis (TBNAT). The enzyme catalyses the acetylation and propionylation of arylamines and hydrazines. Hydralazine is a good acetyl and propionyl acceptor for both MMNAT and TBNAT. The MMNAT structure has been solved to 2.1 Å resolution following crystallisation in the presence of hydralazine and is compared to available NAT structures. From the mode of ligand binding, features of the binding pocket can be identified, which point to a novel mechanism for the acetylation reaction that results in a 3-methyltriazolo[3,4-a]phthalazine ring compound as product.


Subject(s)
Arylamine N-Acetyltransferase/chemistry , Mycobacterium marinum/enzymology , Acetyltransferases/metabolism , Arylamine N-Acetyltransferase/genetics , Arylamine N-Acetyltransferase/metabolism , Catalysis , Catalytic Domain , Crystallization , Mycobacterium/enzymology , Mycobacterium/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Protein Binding
17.
Protein J ; 28(6): 281-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19636684

ABSTRACT

Arylamine N-acetyltansferase (NAT) from Mycobacterium tuberculosis (TBNAT) is a potential drug target for anti-tubercular therapy. Recombinant TBNAT is much less soluble and is produced in lower yields than the closely related NAT from Mycobacterium marinum (MMNAT). In order to explore MMNAT as a model for TBNAT in drug discovery, we compare the two mycobacterial NAT enzymes. Two site-directed mutants of MMNAT have been prepared and characterised: MMNAT71, Tyr --> Phe and MMNAT209, Met --> Thr, in which residues within 6 A of the active-site cysteine have been replaced with the corresponding residue from TBNAT. Two chimeric proteins have also been produced in which the third domain of MMNAT has been replaced by the third domain of TBNAT and vice versa. The activity profile of the chimeric proteins suggests a role for the third domain in the evolutionary divergence of NAT between these closely related mycobacterial species.


Subject(s)
Arylamine N-Acetyltransferase/chemistry , Arylamine N-Acetyltransferase/genetics , Mycobacterium/enzymology , Amino Acid Sequence , Arylamine N-Acetyltransferase/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenylacetates/metabolism , Protein Structure, Tertiary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...