Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
Mol Divers ; 25(3): 1361-1373, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34264440

ABSTRACT

Trypanosomatid-caused diseases are among the neglected infectious diseases with the highest disease burden, affecting about 27 million people worldwide and, in particular, socio-economically vulnerable populations. Trypanothione synthetase (TryS) is considered one of the most attractive drug targets within the thiol-polyamine metabolism of typanosomatids, being unique, essential and druggable. Here, we have compiled a dataset of 401 T. brucei TryS inhibitors that includes compounds with inhibitory data reported in the literature, but also in-house acquired data. QSAR classifiers were derived and validated from such dataset, using publicly available and open-source software, thus assuring the portability of the obtained models. The performance and robustness of the resulting models were substantially improved through ensemble learning. The performance of the individual models and the model ensembles was further assessed through retrospective virtual screening campaigns. At last, as an application example, the chosen model-ensemble has been applied in a prospective virtual screening campaign on DrugBank 5.1.6 compound library. All the in-house scripts used in this study are available on request, whereas the dataset has been included as supplementary material.


Subject(s)
Amide Synthases/chemistry , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Machine Learning , Algorithms , Amide Synthases/antagonists & inhibitors , Amide Synthases/metabolism , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Databases, Pharmaceutical , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Enzyme Inhibitors/pharmacology , Humans , Metabolic Networks and Pathways , Models, Theoretical , ROC Curve , Structure-Activity Relationship
2.
Int J Biol Macromol ; 165(Pt B): 2349-2362, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33098904

ABSTRACT

NAD synthetase (NadE) catalyzes the last step in NAD biosynthesis, transforming deamido-NAD+ into NAD+ by a two-step reaction with co-substrates ATP and amide donor ammonia. In this study, we report the crystal structure of Staphylococcus aureus NAD synthetase enzyme (saNadE) at 2.3 Å resolution. We used this structure to perform molecular dynamics simulations of apo-enzyme, enzyme-substrate (NadE with ATP and NaAD) and enzyme-intermediate complexes (NadE with NaAD-AMP) to investigate key binding interactions and explore the conformational transitions and flexibility of the binding pocket. Our results show large shift of N-terminal region in substrate bound form which is important for ATP binding. Substrates drive the correlated movement of loop regions surrounding it as well as some regions distal to the active site and stabilize them at complex state. Principal component analysis of atomic projections distinguish feasible trajectories to delineate distinct motions in enzyme-substrate to enzyme-intermediate states. Our results suggest mixed binding involving dominant induced fit and conformational selection. MD simulation extracted ensembles of NadE could potentially be utilized for in silico screening and structure based design of more effective Methicillin Resistant Staphylococcus aureus (MRSA) inhibitors.


Subject(s)
Amide Synthases/chemistry , Crystallography, X-Ray , Methicillin-Resistant Staphylococcus aureus/enzymology , Molecular Dynamics Simulation , Apoenzymes/chemistry , Catalytic Domain , Enzyme Stability , Humans , Hydrogen Bonding , NAD/biosynthesis , Principal Component Analysis , Protein Conformation , Protein Subunits/chemistry , Substrate Specificity
3.
Curr Opin Chem Biol ; 55: 77-85, 2020 04.
Article in English | MEDLINE | ID: mdl-32058241

ABSTRACT

The amide functional group is ubiquitous in nature and one of the most important motifs in pharmaceuticals, agrochemicals, and other valuable products. While coupling amides and carboxylic acids is a trivial synthetic transformation, it often requires protective group manipulation, along with stoichiometric quantities of expensive and deleterious coupling reagents. Nature has evolved a range of enzymes to construct amide bonds, the vast majority of which utilize adenosine triphosphate to activate the carboxylic acid substrate for amine coupling. Despite the fact that these enzymes operate under mild conditions, as well as possessing chemoselectivity and regioselectivity that obviates the need for protecting groups, their synthetic potential has been largely unexplored. In this review, we discuss recent research into the discovery, characterization, and development of amide bond forming enzymes, with an emphasis on stand-alone ligase enzymes that can generate amides directly from simple carboxylic acid and amine substrates.


Subject(s)
Amide Synthases/chemistry , Amide Synthases/metabolism , Amides/chemistry , Acyltransferases/metabolism , Adenosine Triphosphate/metabolism , Amines/chemistry , Biocatalysis , Carboxylic Acids/chemistry , Coenzyme A/metabolism , Peptide Synthases/metabolism , Protein Conformation , Substrate Specificity
4.
Nat Commun ; 11(1): 16, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31911602

ABSTRACT

NAD+ synthetase is an essential enzyme of de novo and recycling pathways of NAD+ biosynthesis in Mycobacterium tuberculosis but not in humans. This bifunctional enzyme couples the NAD+ synthetase and glutaminase activities through an ammonia tunnel but free ammonia is also a substrate. Here we show that the Homo sapiens NAD+ synthetase (hsNadE) lacks substrate specificity for glutamine over ammonia and displays a modest activation of the glutaminase domain compared to tbNadE. We report the crystal structures of hsNadE and NAD+ synthetase from M. tuberculosis (tbNadE) with synthetase intermediate analogues. Based on the observed exclusive arrangements of the domains and of the intra- or inter-subunit tunnels we propose a model for the inter-domain communication mechanism for the regulation of glutamine-dependent activity and NH3 transport. The structural and mechanistic comparison herein reported between hsNadE and tbNadE provides also a starting point for future efforts in the development of anti-TB drugs.


Subject(s)
Amide Synthases/metabolism , Ammonia/metabolism , Bacterial Proteins/metabolism , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Mycobacterium tuberculosis/enzymology , Amide Synthases/chemistry , Amide Synthases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/chemistry , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Catalytic Domain , Glutaminase/chemistry , Glutaminase/genetics , Glutaminase/metabolism , Glutamine/metabolism , Humans , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , NAD/metabolism , Substrate Specificity
5.
Protein Pept Lett ; 26(5): 371-376, 2019.
Article in English | MEDLINE | ID: mdl-30827222

ABSTRACT

BACKGROUND: Leishmaniasis is caused by a protozoan parasite, Leishmania. It is common in more than 98 countries throughout the world. Due to insufficient availability of antileishmanial chemotherapeutics, it is an urgent need to search for new molecules which have better efficacy, low toxicity and are available at low cost. OBJECTIVES: There is a high rate of diabetic cases throughout the world that is why we planned to test the antileishmanial activity of glyburide, an effective sugar lowering drug used for the treatment of diabetes. In this study, glyburide showed a significant decrease in the parasite growth and survival in vitro in a dose-dependent manner. METHODS: Anti-leishmanial activity of glyburide was checked by culturing Leishmania donovani promastigotes in the presence of glyburide in a dose and time dependent manner. Docking study against Leishmania donovani-Trypanothione synthetase (LdTrySyn) protein was performed using Autodock Vina tool. RESULTS: Growth reversibility assay shows that growth of treated parasite was not reversed when transferred to fresh culture media after 7 days. Moreover, docking studies show efficient interactions of glyburide with key residues in the catalytic site of Leishmania donovani- Trypanothione synthetase (LdTrySyn), a very important leishmanial enzyme involved in parasite's survival by detoxification of Nitric Oxide (NO) species, generated by the mammalian host as a defense molecule. Thus this study proves that the drug-repurposing is a beneficial strategy for identification of new and potent antileishmanial molecules. CONCLUSION: The results suggest that glyburide binds to LdTrySyn and inhibits its activity which further leads to the altered parasite morphology and inhibition of parasite growth. Glyburide may also be used in combination with other anti-leishmanial drugs to potentiate the response of the chemotherapy. Overall this study provides information about combination therapy as well as a single drug treatment for the infected patients suffering from diabetes. This study also provides raw information for further in vivo disease model studies to confirm the hypothesis.


Subject(s)
Antiprotozoal Agents/pharmacology , Glyburide/pharmacology , Leishmania donovani/drug effects , Leishmaniasis/drug therapy , Amide Synthases/chemistry , Antiprotozoal Agents/therapeutic use , Catalytic Domain , Drug Repositioning , Glyburide/therapeutic use , Humans , Molecular Docking Simulation , Protozoan Proteins/chemistry
6.
J Glob Antimicrob Resist ; 18: 187-194, 2019 09.
Article in English | MEDLINE | ID: mdl-30797085

ABSTRACT

OBJECTIVES: Green tea contains a predominant set of polyphenolic compounds with biological activities. The aim of this study was to investigate the antileishmanial activities of the main components of green tea, including catechin, (-)-epicatechin, epicatechin gallate (ECG) and (-)-epigallocatechin 3-O-gallate (EGCG), against Leishmania infantum promastigotes. METHODS: Green tea ligands and the control drug pentamidine were docked using AutoDock 4.3 software into the active sites of trypanothione synthetase and arginase, which were modelled using homology modelling programs. The colorimetric MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay was used to measure L. infantum promastigotes at different concentrations of green tea compounds in a concentration- and time-dependent manner. Results were expressed as 50% and 90% inhibitory concentrations (IC50 and IC90, respectively). RESULTS: In silico and in vitro assays showed that all of the green tea compounds have antileishmanial activity. EGCG and ECG were the most active compounds against L. infantum promastigotes, with IC50 values of 27.7µM and 75µM and IC90 values of 88.4µM and 188.7µM, respectively. Pentamidine displayed greater growth inhibition than all of the other tested compounds in a concentration- and time-dependent manner. CONCLUSION: In this study, in silico and docking results were in accordance with the in vitro activity of the compounds. Moreover, EGCG and ECG showed reasonable levels of selectivity for Leishmania.


Subject(s)
Leishmania infantum/drug effects , Plant Exudates/pharmacology , Tea/chemistry , Amide Synthases/chemistry , Amide Synthases/drug effects , Antioxidants/pharmacology , Arginase/chemistry , Arginase/drug effects , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Computer Simulation , Iran , Leishmaniasis, Visceral/parasitology , Microbial Sensitivity Tests , Molecular Docking Simulation , Pentamidine/chemistry , Pentamidine/pharmacology , Protease Inhibitors/pharmacology
7.
J Biol Chem ; 293(19): 7397-7407, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29581233

ABSTRACT

NADH (NAD+) and its reduced form NADH serve as cofactors for a variety of oxidoreductases that participate in many metabolic pathways. NAD+ also is used as substrate by ADP-ribosyl transferases and by sirtuins. NAD+ biosynthesis is one of the most fundamental biochemical pathways in nature, and the ubiquitous NAD+ synthetase (NadE) catalyzes the final step in this biosynthetic route. Two different classes of NadE have been described to date: dimeric single-domain ammonium-dependent NadENH3 and octameric glutamine-dependent NadEGln, and the presence of multiple NadE isoforms is relatively common in prokaryotes. Here, we identified a novel dimeric group of NadEGln in bacteria. Substrate preferences and structural analyses suggested that dimeric NadEGln enzymes may constitute evolutionary intermediates between dimeric NadENH3 and octameric NadEGln The characterization of additional NadE isoforms in the diazotrophic bacterium Azospirillum brasilense along with the determination of intracellular glutamine levels in response to an ammonium shock led us to propose a model in which these different NadE isoforms became active accordingly to the availability of nitrogen. These data may explain the selective pressures that support the coexistence of multiple isoforms of NadE in some prokaryotes.


Subject(s)
Adaptation, Physiological , Azospirillum brasilense/enzymology , Biological Evolution , Glutamine/metabolism , Herbaspirillum/enzymology , Mycobacterium tuberculosis/enzymology , Amide Synthases/chemistry , Amide Synthases/metabolism , Amino Acid Sequence , Ammonia/metabolism , Azospirillum brasilense/metabolism , Azospirillum brasilense/physiology , Catalysis , Herbaspirillum/metabolism , Herbaspirillum/physiology , Kinetics , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/physiology , NAD/metabolism , Phylogeny , Protein Multimerization , Sequence Homology, Amino Acid , Substrate Specificity
8.
FEBS Lett ; 591(23): 3881-3894, 2017 12.
Article in English | MEDLINE | ID: mdl-29127710

ABSTRACT

Buthionine sulfoximine (BSO) induces decreased glutathione (GSH) and trypanothione [T(SH)2 ] pools in trypanosomatids, presumably because only gamma-glutamylcysteine synthetase (γECS) is blocked. However, some BSO effects cannot be explained by exclusive γECS inhibition; therefore, its effect on the T(SH)2 metabolism pathway in Trypanosoma cruzi was re-examined. Parasites exposed to BSO did not synthesize T(SH)2 even when supplemented with cysteine or GSH, suggesting trypanothione synthetase (TryS) inhibition by BSO. Indeed, recombinant γECS and TryS, but not GSH synthetase, were inhibited by BSO and kinetics and docking analyses on a TcTryS 3D model suggested BSO binding at the GSH site. Furthermore, parasites overexpressing γECS and TryS showed ~ 50% decreased activities after BSO treatment. These results indicated that BSO is also an inhibitor of TryS.


Subject(s)
Buthionine Sulfoximine/pharmacology , Glutathione/analogs & derivatives , Spermidine/analogs & derivatives , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/metabolism , Amide Synthases/antagonists & inhibitors , Amide Synthases/chemistry , Amide Synthases/genetics , Animals , Enzyme Inhibitors/pharmacology , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/genetics , Glutathione/biosynthesis , Glutathione/metabolism , Glutathione Synthase/antagonists & inhibitors , Glutathione Synthase/genetics , Humans , Kinetics , Metabolic Networks and Pathways/drug effects , Molecular Docking Simulation , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spermidine/biosynthesis , Trypanosoma cruzi/genetics
9.
FEBS J ; 284(15): 2425-2441, 2017 08.
Article in English | MEDLINE | ID: mdl-28618168

ABSTRACT

Streptococcus pyogenes, also known as Group A Strep (GAS), is an obligate human pathogen that is responsible for millions of infections and numerous deaths per year. Infection manifestations can range from simple, acute pharyngitis to more complex, necrotizing fasciitis. To date, most treatments for GAS infections involve the use of common antibiotics including tetracycline and clindamycin. Unfortunately, new strains have been identified that are resistant to these drugs, therefore, new targets must be identified to treat drug-resistant strains. This work is focused on the structural and functional characterization of three proteins: spNadC, spNadD, and spNadE. These enzymes are involved in the biosynthesis of nicotinamide adenine dinucleotide (NAD+ ). The structures of spNadC and spNadE were determined. SpNadC is suggested to play a role in GAS virulence, while spNadE, functions as an NAD synthetase and is considered to be a new drug target. Determination of the spNadE structure uncovered a putative, NH3 channel, which may provide insight into the mechanistic details of NH3 -dependent NAD+ synthetases in prokaryotes. ENZYMES: Quinolinate phosphoribosyltransferase: EC2.4.2.19 and NAD synthetase: EC6.3.1.5. DATABASE: Protein structures for spNadC, spNadCΔ69A , and spNadE are deposited into Protein Data Bank under the accession codes 5HUL, 5HUO & 5HUP, and 5HUH & 5HUJ, respectively.


Subject(s)
Amide Synthases/metabolism , Bacterial Proteins/metabolism , Models, Molecular , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Pentosyltransferases/metabolism , Quinolinic Acid/metabolism , Streptococcus pyogenes/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amide Synthases/chemistry , Amide Synthases/genetics , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Catalytic Domain , Cluster Analysis , Computational Biology , Crystallography, X-Ray , Dimerization , Gene Deletion , Nicotinamide-Nucleotide Adenylyltransferase/chemistry , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Pentosyltransferases/chemistry , Pentosyltransferases/genetics , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structural Homology, Protein
10.
Angew Chem Int Ed Engl ; 56(21): 5937-5941, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28440038

ABSTRACT

The antilarval mushroom polyenes 18-methyl-19-oxoicosaoctaenoic acid and 20-methyl-21-oxodocosanonaenoic acid appear in response to injury of the mycelium of the stereaceous mushroom BY1. We identified a polyketide synthase (PPS1) which belongs to a hitherto completely uncharacterized clade of polyketide synthases. Expression of the PPS1 gene is massively upregulated following mycelial damage. The synthesis of the above polyenes was reconstituted in the mold Aspergillus niger as a heterologous host. This demonstrates that PPS1 1) synchronously produces branched-chain polyketides of varied lengths, and 2) catalyzes the unprecedented shift of eight or nine double bonds. This study represents the first characterization of a reducing polyketide synthase from a mushroom. We also show that injury-induced de novo synthesis of polyketides is a fungal response strategy.


Subject(s)
Agaricales , Polyenes/chemistry , Amide Synthases/chemistry , Host-Parasite Interactions , Molecular Structure
11.
Org Biomol Chem ; 14(19): 4534-41, 2016 May 11.
Article in English | MEDLINE | ID: mdl-27101411

ABSTRACT

Herein we report a practical synthetic route to the lasso peptide lassomycin () and C-terminal variant lassomycin-amide (). The biological evaluation of peptides and against Mycobacterium tuberculosis revealed that neither had any activity against this bacterium. This lack of biological activity has led us to propose that naturally occurring lassomycin may actually exhibit a standard lasso peptide threaded conformation rather than the previously reported unthreaded structure.


Subject(s)
Amide Synthases/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/chemical synthesis , Amino Acid Sequence , Chemistry Techniques, Synthetic , Molecular Conformation , Protein Conformation
12.
J Comput Aided Mol Des ; 30(4): 305-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26891837

ABSTRACT

Chagas disease is a parasitic infection caused by the protozoa Trypanosoma cruzi that affects about 6 million people in Latin America. Despite its sanitary importance, there are currently only two drugs available for treatment: benznidazole and nifurtimox, both exhibiting serious adverse effects and limited efficacy in the chronic stage of the disease. Polyamines are ubiquitous to all living organisms where they participate in multiple basic functions such as biosynthesis of nucleic acids and proteins, proliferation and cell differentiation. T. cruzi is auxotroph for polyamines, which are taken up from the extracellular medium by efficient transporters and, to a large extent, incorporated into trypanothione (bis-glutathionylspermidine), the major redox cosubstrate of trypanosomatids. From a 268-compound database containing polyamine analogs with and without inhibitory effect on T. cruzi we have inferred classificatory models that were later applied in a virtual screening campaign to identify anti-trypanosomal compounds among drugs already used for other therapeutic indications (i.e. computer-guided drug repositioning) compiled in the DrugBank and Sweetlead databases. Five of the candidates identified with this strategy were evaluated in cellular models from different pathogenic trypanosomatids (T. cruzi wt, T. cruzi PAT12, T. brucei and Leishmania infantum), and in vitro models of aminoacid/polyamine transport assays and trypanothione synthetase inhibition assay. Triclabendazole, sertaconazole and paroxetine displayed inhibitory effects on the proliferation of T. cruzi (epimastigotes) and the uptake of putrescine by the parasite. They also interfered with the uptake of others aminoacids and the proliferation of infective T. brucei and L. infantum (promastigotes). Trypanothione synthetase was ruled out as molecular target for the anti-parasitic activity of these compounds.


Subject(s)
Amide Synthases/antagonists & inhibitors , Chagas Disease/drug therapy , Drug Repositioning , Polyamines/chemistry , Amide Synthases/chemistry , Antiprotozoal Agents/chemistry , Chagas Disease/parasitology , Computer Simulation , Glutathione/analogs & derivatives , Glutathione/chemistry , Glutathione/therapeutic use , Humans , Imidazoles/chemistry , Imidazoles/therapeutic use , Nitroimidazoles/chemistry , Nitroimidazoles/therapeutic use , Polyamines/therapeutic use , Spermidine/analogs & derivatives , Spermidine/chemistry , Spermidine/therapeutic use , Thiophenes/chemistry , Thiophenes/therapeutic use , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/pathogenicity , User-Computer Interface
13.
Sci Rep ; 5: 12744, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26244829

ABSTRACT

Nosiheptide is a parent compound of thiopeptide family that exhibit potent activities against various bacterial pathogens. Its C-terminal amide formation is catalyzed by NosA, which is an unusual strategy for maturating certain thiopeptides by processing their precursor peptides featuring a serine extension. We here report the crystal structure of truncated NosA1-111 variant, revealing three key elements, including basic lysine 49 (K49), acidic glutamic acid 101 (E101) and flexible C-terminal loop NosA112-151, are crucial to the catalytic terminal amide formation in nosiheptide biosynthesis. The side-chain of residue K49 and the C-terminal loop fasten the substrate through hydrogen bonds and hydrophobic interactions. The side-chain of residue E101 enhances nucleophilic attack of H2O to the methyl imine intermediate, leading to Cα-N bond cleavage and nosiheptide maturation. The sequence alignment of NosA and its homologs NocA, PbtH, TpdK and BerI, and the enzymatic assay suggest that the mechanistic studies on NosA present an intriguing paradigm about how NosA family members function during thiopeptide biosynthesis.


Subject(s)
Amide Synthases/chemistry , Crystallography, X-Ray , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Thiazoles/chemistry
14.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 6): 763-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26057809

ABSTRACT

Staphylococcus aureus is an important human and animal pathogen that causes a wide range of infections. The prevalence of multidrug-resistant S. aureus strains in both hospital and community settings makes it imperative to characterize new drug targets to combat S. aureus infections. In this context, enzymes involved in NAD metabolism and synthesis are significant drug targets as NAD is a central player in several cellular processes. NAD synthetase catalyzes the last step in the biosynthesis of nicotinamide adenine dinucleotide, making it a crucial intermediate enzyme linked to the biosynthesis of several amino acids, purine and pyrimidine nucleotides, coenzymes and antibiotics.


Subject(s)
Amide Synthases/chemistry , Bacterial Proteins/chemistry , Methicillin-Resistant Staphylococcus aureus/chemistry , NAD/chemistry , Amide Synthases/genetics , Amide Synthases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Metabolic Networks and Pathways , Methicillin-Resistant Staphylococcus aureus/enzymology , Molecular Sequence Data , NAD/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , X-Ray Diffraction
15.
PLoS One ; 9(6): e97600, 2014.
Article in English | MEDLINE | ID: mdl-24901644

ABSTRACT

Kinetoplastids differ from other organisms in their ability to conjugate glutathione and spermidine to form trypanothione which is involved in maintaining redox homeostasis and removal of toxic metabolites. It is also involved in drug resistance, antioxidant mechanism, and defense against cellular oxidants. Trypanothione synthetase (TryS) of thiol metabolic pathway is the sole enzyme responsible for the biosynthesis of trypanothione in Leishmania donovani. In this study, TryS gene of L. donovani (LdTryS) was cloned, expressed, and fusion protein purified with affinity column chromatography. The purified protein showed optimum enzymatic activity at pH 8.0-8.5. The TryS amino acids sequences alignment showed that all amino acids involved in catalytic and ligands binding of L. major are conserved in L. donovani. Subcellular localization using digitonin fractionation and immunoblot analysis showed that LdTryS is localized in the cytoplasm. Furthermore, RT-PCR coupled with immunoblot analysis showed that LdTryS is overexpressed in Amp B resistant and stationary phase promastigotes (∼ 2.0-folds) than in sensitive strain and logarithmic phase, respectively, which suggests its involvement in Amp B resistance. Also, H2O2 treatment upto 150 µM for 8 hrs leads to 2-fold increased expression of LdTryS probably to cope up with oxidative stress generated by H2O2. Therefore, this study demonstrates stage- and Amp B sensitivity-dependent expression of LdTryS in L. donovani and involvement of TryS during oxidative stress to help the parasites survival.


Subject(s)
Amide Synthases/genetics , Amphotericin B/pharmacology , Drug Resistance/genetics , Gene Expression Regulation/drug effects , Leishmania donovani/drug effects , Leishmania donovani/genetics , Leishmaniasis, Visceral/parasitology , Amide Synthases/chemistry , Amide Synthases/isolation & purification , Amide Synthases/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Enzyme Activation , Gene Expression , Humans , Hydrogen Peroxide/metabolism , Leishmania donovani/growth & development , Leishmaniasis, Visceral/drug therapy , Life Cycle Stages , Molecular Sequence Data , Oxidation-Reduction , Parasitic Sensitivity Tests , Protein Multimerization , Protein Transport , Reactive Oxygen Species/metabolism , Sequence Alignment , Up-Regulation
16.
J Am Chem Soc ; 134(3): 1673-9, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22136518

ABSTRACT

The amide synthase of the geldanamycin producer, Streptomyces hygroscopicus, shows a broader chemoselectivity than the corresponding amide synthase present in Actinosynnema pretiosum, the producer of the highly cytotoxic ansamycin antibiotics, the ansamitocins. This was demonstrated when blocked mutants of both strains incapable of biosynthesizing 3-amino-5-hydroxybenzoic acid (AHBA), the polyketide synthase starter unit of both natural products, were supplemented with 3-amino-5-hydroxymethylbenzoic acid instead. Unlike the ansamitocin producer A. pretiosum, S. hygroscopicus processed this modified starter unit not only to the expected 19-membered macrolactams but also to ring enlarged 20-membered macrolactones. The former mutaproducts revealed the sequence of transformations catalyzed by the post-PKS tailoring enzymes in geldanamycin biosynthesis. The unprecedented formation of the macrolactones together with molecular modeling studies shed light on the mode of action of the amide synthase responsible for macrocyclization. Obviously, the 3-hydroxymethyl substituent shows similar reactivity and accessibility toward C-1 of the seco-acid as the arylamino group, while phenolic hydroxyl groups lack this propensity to act as nucleophiles in the macrocyclization. The promiscuity of the amide synthase of S. hygroscopicus was further demonstrated by successful feeding of four other m-hydroxymethylbenzoic acids, leading to formation of the expected 20-membered macrocycles. Good to moderate antiproliferative activities were encountered for three of the five new geldanamycin derivatives, which matched well with a competition assay for Hsp90α.


Subject(s)
Amide Synthases/metabolism , Benzoquinones/metabolism , Lactams, Macrocyclic/metabolism , Streptomyces/enzymology , Amide Synthases/chemistry , Amino Acid Sequence , Benzoquinones/chemistry , Lactams, Macrocyclic/chemistry , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Streptomyces/chemistry , Substrate Specificity
17.
Wei Sheng Wu Xue Bao ; 51(3): 305-12, 2011 Mar.
Article in Chinese | MEDLINE | ID: mdl-21604544

ABSTRACT

Nicotinamide-adenine dinucleotide (phosphate) (NAD(P)) metabolism involves many fundamental cellular events, such as energy metabolism, maintenance of redox homeostasis and regulation of cell longevity. Inhibitors of essential enzymes of NAD(P) biosynthetic pathways might be promising leads for novel antibiotics, such as the NAD synthase inhibitors. This review described the crystal structural, functional properties, regulator and structure-based inhibitors design for NAD synthase. This might provide the basis for developing NAD-based therapeutics.


Subject(s)
Amide Synthases/antagonists & inhibitors , Amide Synthases/metabolism , Enzyme Inhibitors/pharmacology , Amide Synthases/chemistry , Amino Acid Sequence , Enzyme Inhibitors/chemistry , Molecular Sequence Data
18.
Protein Sci ; 20(3): 557-66, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21226054

ABSTRACT

The bifunctional Escherichia coli glutathionylspermidine synthetase/amidase (GspSA) catalyzes both the synthesis and hydrolysis of Gsp. Its amidase domain (GspA), which catalyzes the hydrolysis of Gsp into glutathione and spermidine, plays an important role in redox sensing and protein S-thiolation. To gain insight of the regulation and catalytic mechanism of and further understand the recycling of the Gsp dimer and Gsp-S-protein adducts, we solved two crystal structures of GspA and GspSA both with the C59A mutation and bound with the substrate, Gsp. In both structures, Cys59, His131, and Glu147 form the catalytic triad, which is similar to other cysteine proteases. Comparison of the GspA_Gsp complex and apo GspSA structures indicates that on binding with Gsp, the side chains of Asn149 and Gln58 of the amidase domain are induced to move closer to the carbonyl oxygen of the cleaved amide bond of Gsp, thereby participating in catalysis. In addition, the helix-loop region of GspA, corresponding to the sequence (30)YSSLDPQEYEDDA(42), involves in regulating the substrate binding. Our previous study indicated that the thiol of Cys59 of GspA is only oxidized to sulfenic acid by H(2)O(2). When comparing the active site of GspA with those of other cysteine proteases, we found that limited space and hydrophobicity of the environment around Cys59 play an important role to inhibit its further oxidation. The structural results presented here not only elucidate the catalytic mechanism and regulation of GspA but also help us to design small molecules to inhibit or probe for the activity of GspA.


Subject(s)
Amide Synthases/chemistry , Amidohydrolases/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Protein Conformation , Amide Synthases/genetics , Amide Synthases/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Amino Acid Sequence , Catalytic Domain , Cysteine/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glutathione/analogs & derivatives , Glutathione/chemistry , Glutathione/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Oxidation-Reduction , Protein Binding , Sequence Alignment , Spermidine/analogs & derivatives , Spermidine/chemistry , Spermidine/metabolism
19.
J Biol Chem ; 285(50): 38853-60, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20926388

ABSTRACT

Fredericamycin (FDM) A is a pentadecaketide natural product that features an amide linkage. Analysis of the fdm cluster from Streptomyces griseus ATCC 43944, however, failed to reveal genes encoding the types of amide synthetases commonly seen in natural product biosynthesis. Here, we report in vivo and in vitro characterizations of FdmV, an asparagine synthetase (AS) B-like protein, as an amide synthetase that catalyzes the amide bond formation in FDM A biosynthesis. This is supported by the findings that (i) inactivation of fdmV in vivo afforded the ΔfdmV mutant strain SB4027 that abolished FDM A and FDM E production but accumulated FDM C, a biosynthetic intermediate devoid of the characteristic amide linkage; (ii) FdmV in vitro catalyzes conversion of FDM C to FDM B, a known intermediate for FDM A biosynthesis (apparent K(m) = 162 ± 67 µM and k(cat) = 0.11 ± 0.02 min(-1)); and (iii) FdmV also catalyzes the amidation of FDM M-3, a structural analog of FDM C, to afford amide FDM M-6 in vitro, albeit at significantly reduced efficiency. Preliminary enzymatic studies revealed that, in addition to the common nitrogen sources (L-Gln and free amine) of class II glutamine amidotransferases (to which AS B belongs), FdmV can also utilize L-Asn as a nitrogen donor. The amide bond formation in FDM A biosynthesis is proposed to occur after C-8 hydroxylation but before the carbaspirocycle formation.


Subject(s)
Amide Synthases/chemistry , Amides/chemistry , Gene Expression Regulation, Bacterial , Streptomyces griseus/metabolism , Asparagine/chemistry , Aspartate-Ammonia Ligase/chemistry , Catalysis , Catalytic Domain , Hydroxylation , Isoquinolines/metabolism , Kinetics , Models, Chemical , Mutation , Nitrogen/chemistry , Recombinant Proteins/chemistry , Spiro Compounds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...