Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38673904

ABSTRACT

Chagas disease is one of the world's neglected tropical diseases, caused by the human pathogenic protozoan parasite Trypanosoma cruzi. There is currently a lack of effective and tolerable clinically available therapeutics to treat this life-threatening illness and the discovery of modern alternative options is an urgent matter. T. cruzi glucokinase (TcGlcK) is a potential drug target because its product, d-glucose-6-phosphate, serves as a key metabolite in the pentose phosphate pathway, glycolysis, and gluconeogenesis. In 2019, we identified a novel cluster of TcGlcK inhibitors that also exhibited anti-T. cruzi efficacy called the 3-nitro-2-phenyl-2H-chromene analogues. This was achieved by performing a target-based high-throughput screening (HTS) campaign of 13,040 compounds. The selection criteria were based on first determining which compounds strongly inhibited TcGlcK in a primary screen, followed by establishing on-target confirmed hits from a confirmatory assay. Compounds that exhibited notable in vitro trypanocidal activity over the T. cruzi infective form (trypomastigotes and intracellular amastigotes) co-cultured in NIH-3T3 mammalian host cells, as well as having revealed low NIH-3T3 cytotoxicity, were further considered. Compounds GLK2-003 and GLK2-004 were determined to inhibit TcGlcK quite well with IC50 values of 6.1 µM and 4.8 µM, respectively. Illuminated by these findings, we herein screened a small compound library consisting of thirteen commercially available 3-nitro-2-phenyl-2H-chromene analogues, two of which were GLK2-003 and GLK2-004 (compounds 1 and 9, respectively). Twelve of these compounds had a one-point change from the chemical structure of GLK2-003. The analogues were run through a similar primary screening and confirmatory assay protocol to our previous HTS campaign. Subsequently, three in vitro biological assays were performed where compounds were screened against (a) T. cruzi (Tulahuen strain) infective form co-cultured within NIH-3T3 cells, (b) T. brucei brucei (427 strain) bloodstream form, and (c) NIH-3T3 host cells alone. We report on the TcGlcK inhibitor constant determinations, mode of enzyme inhibition, in vitro antitrypanosomal IC50 determinations, and an assessment of structure-activity relationships. Our results reveal that the 3-nitro-2-phenyl-2H-chromene scaffold holds promise and can be further optimized for both Chagas disease and human African trypanosomiasis early-stage drug discovery research.


Subject(s)
Benzopyrans , Glucokinase , Trypanocidal Agents , Trypanosoma cruzi , Animals , Humans , Mice , Benzopyrans/pharmacology , Benzopyrans/chemistry , Chagas Disease/drug therapy , Chagas Disease/parasitology , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Glucokinase/metabolism , Glucokinase/antagonists & inhibitors , High-Throughput Screening Assays , Molecular Docking Simulation , NIH 3T3 Cells , Structure-Activity Relationship , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemistry , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/enzymology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
2.
Biochimie ; 218: 8-19, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37741546

ABSTRACT

The hole mutagenesis approach was used to interrogate the importance of F337 in Trypanosoma cruzi glucokinase (TcGlcK) in order to understand the complete set of binding interactions that are made by d-glucosamine analogue inhibitors containing aromatic tail groups that can extend to the outer part of the active site. An interesting inhibitor of this analogue class includes 2-N-carboxybenzyl-2-deoxy-d-glucosamine (CBZ-GlcN), which exhibits strong TcGlcK binding with a Ki of 710 nM. The residue F337 is found at the outer part of the active site that stems from the second protein subunit of the homodimeric assembly. In this study, F337 was changed to leucine and alanine so as to diminish phenylalanine's side chain size and attenuate intermolecular interactions in this region of the binding cavity. Results from enzyme - inhibitor assays revealed that the phenyl group of F337 made dominant hydrophobic interactions with the phenyl group of CBZ-GlcN as opposed to π - π stacking interactions. Moreover, enzymatic activity assays and X-ray crystallographic experiments indicated that each of these site-directed mutants primarily retained their activity and had high structural similarity of their protein fold. A computed structure model of T. cruzi hexokinase (TcHxK), which was produced by the artificial intelligence system AlphaFold, was compared to an X-ray crystal structure of TcGlcK. Our structural analysis revealed that TcHxK lacked an F337 counterpart residue and probably exists in the monomeric form. We proposed that the d-glucosamine analogue inhibitors that are structurally similar to CBZ-GlcN may not bind as strongly in TcHxK as they do in TcGlcK because of absent van der Waals contact from residue side chains.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Humans , Glucokinase/chemistry , Glucokinase/metabolism , Catalytic Domain , Phenylalanine , Artificial Intelligence , Models, Molecular , Glucosamine , Binding Sites , Crystallography, X-Ray
3.
Bioorg Med Chem Lett ; 47: 128227, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34174398

ABSTRACT

Eighteen amino sugar analogues were screened against Trypanosoma cruzi glucokinase (TcGlcK), a potential drug-target of the protozoan parasite in order to assess for viable enzyme inhibition. The analogues were divided into three amino sugar scaffolds that included d-glucosamine (d-GlcN), d-mannosamine (d-ManN), and d-galactosamine (d-GalN); moreover, all but one of these compounds were novel. TcGlcK is an important metabolic enzyme that has a role in producing G6P for glycolysis and the pentose phosphate pathway (PPP). The inhibition of these pathways via glucose kinases (i.e., glucokinase and hexokinase) appears to be a strategic approach for drug discovery. Glucose kinases phosphorylate d-glucose with co-substrate ATP to yield G6P and the formed G6P enters both pathways for catabolism. The compound screen revealed five on-target confirmed inhibitors that were all from the d-GlcN series, such as compounds 1, 2, 4, 5, and 6. Four of these compounds were strong TcGlcK inhibitors (1, 2, 4, and 6) since they were found to have micromolar inhibitory constant (Ki) values around 20 µM. Three of the on-target confirmed inhibitors (1, 5, and 6) revealed notable in vitro anti-T. cruzi activity with IC50 values being less than 50 µM. Compound 1 was benzoyl glucosamine (BENZ-GlcN), a known TcGlcK inhibitor that was the starting point for the design of the compounds in this study; in addition, TcGlcK - compound 1 inhibition properties were previously determined [D'Antonio, E. L. et al. (2015) Mol. Biochem. Parasitol. 204, 64-76]. As such, compounds 5 and 6 were further evaluated biochemically, where formal Ki values were determined as well as their mode of TcGlcK inhibition. The Ki values determined for compounds 5 and 6 were 107 ± 4 µM and 15.2 ± 3.3 µM, respectively, and both of these compounds exhibited the competitive inhibition mode.


Subject(s)
Amino Sugars/pharmacology , Enzyme Inhibitors/pharmacology , Glucokinase/antagonists & inhibitors , Trypanosoma cruzi/enzymology , Amino Sugars/chemical synthesis , Amino Sugars/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glucokinase/metabolism , Molecular Structure , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 32: 127723, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33249135

ABSTRACT

Ribose 5-phosphate isomerase type B (RPI-B) is a key enzyme of the pentose phosphate pathway that catalyzes the isomerization of ribose 5-phosphate (R5P) and ribulose 5-phosphate (Ru5P). Trypanosoma cruzi RPI-B (TcRPI-B) appears to be a suitable drug-target mainly due to: (i) its essentiality (as previously shown in other trypanosomatids), (ii) it does not present a homologue in mammalian genomes sequenced thus far, and (iii) it participates in the production of NADPH and nucleotide/nucleic acid synthesis that are critical for parasite cell survival. In this survey, we report on the competitive inhibition of TcRPI-B by a substrate - analogue inhibitor, Compound B (Ki = 5.5 ± 0.1 µM), by the Dixon method. This compound has an iodoacetamide moiety that is susceptible to nucleophilic attack, particularly by the cysteine thiol group. Compound B was conceived to specifically target Cys-69, an important active site residue. By incubating TcRPI-B with Compound B, a trypsin digestion LC-MS/MS analysis revealed the identification of Compound B covalently bound to Cys-69. This inhibitor also exhibited notable in vitro trypanocidal activity against T. cruzi infective life-stages co-cultured in NIH-3T3 murine host cells (IC50 = 17.40 ± 1.055 µM). The study of Compound B served as a proof-of-concept so that next generation inhibitors can potentially be developed with a focus on using a prodrug group in replacement of the iodoacetamide moiety, thus representing an attractive starting point for the future treatment of Chagas' disease.


Subject(s)
Aldose-Ketose Isomerases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Protozoan Proteins/antagonists & inhibitors , Trypanocidal Agents/chemical synthesis , Trypanosoma cruzi/enzymology , 3T3 Cells , Aldose-Ketose Isomerases/metabolism , Animals , Binding Sites , Catalytic Domain , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Kinetics , Mice , Molecular Dynamics Simulation , Protozoan Proteins/metabolism , Substrate Specificity , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects
5.
Bioorg Med Chem Lett ; 29(15): 1948-1953, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31133533

ABSTRACT

A high-throughput screening (HTS) campaign was carried out for Trypanosoma cruzi glucokinase (TcGlcK), a potential drug-target of the pathogenic protozoan parasite. Glycolysis and the pentose phosphate pathway (PPP) are important metabolic pathways for T. cruzi and the inhibition of the glucose kinases (i.e. glucokinase and hexokinase) may be a strategic approach for drug discovery. Glucose kinases phosphorylate d-glucose with co-substrate ATP to yield G6P, and moreover, the produced G6P enters both pathways for catabolism. The TcGlcK - HTS campaign revealed 25 novel enzyme inhibitors that were distributed in nine chemical classes and were discovered from a primary screen of 13,040 compounds. Thirteen of these compounds were found to have low micromolar IC50 enzyme - inhibition values; strikingly, four of those compounds exhibited low toxicity towards NIH-3T3 murine host cells and notable in vitro trypanocidal activity. These compounds were of three chemical classes: (a) the 3-nitro-2-phenyl-2H-chromene scaffold, (b) the N-phenyl-benzenesulfonamide scaffold, and (c) the gossypol scaffold. Two compounds from the 3-nitro-2-phenyl-2H-chromene scaffold were determined to be hit-to-lead candidates that can proceed further down the early-stage drug discovery process.


Subject(s)
Chagas Disease/drug therapy , Enzyme Inhibitors/therapeutic use , Glucokinase/therapeutic use , High-Throughput Screening Assays/methods , Enzyme Inhibitors/pharmacology , Glucokinase/pharmacology , Trypanosoma cruzi
6.
Mol Biochem Parasitol ; 227: 47-52, 2019 01.
Article in English | MEDLINE | ID: mdl-30571993

ABSTRACT

Glucokinase from pathogenic protozoa of the genus Leishmania is a potential drug target for the chemotherapeutic treatment against leishmaniasis because this enzyme is located at a nodal point between two critically important metabolic pathways, glycolysis and the pentose phosphate pathway (PPP). L. braziliensis glucokinase (LbGlcK) was evaluated for its structural characterization and enzymatic performance. The enzyme catalyzes the phosphorylation of d-glucose with co-substrate ATP to yield the products G6P and ADP. LbGlcK had KM values determined as 6.61 ± 2.63 mM and 0.338 ± 0.080 mM for d-glucose and ATP, respectively. The 1.85 Å resolution X-ray crystal structure of the apo form of LbGlcK was determined and a homodimer was revealed where each subunit (both in open conformations) included the typical small and large domains. Structural comparisons were assessed in relationship to Homo sapiens hexokinase IV and Trypanosoma cruzi glucokinase. Comparisons revealed that all residues important for making hydrogen bonding interactions with d-glucose in the active site and catalysis were strictly conserved. LbGlcK was screened against four glucosamine analogue inhibitors and the stronger inhibitor of the series, HPOP-GlcN, had a Ki value of 56.9 ± 16.6 µM that exhibited competitive inhibition. For the purpose of future structure-based drug design experimentation, L. braziliensis glucokinase was observed to be very similar to T. cruzi glucokinase even though there was a 44% protein sequence identity between the two enzymes.


Subject(s)
Glucokinase/chemistry , Glucokinase/metabolism , Leishmania braziliensis/enzymology , Leishmaniasis, Cutaneous/parasitology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Glucokinase/genetics , Glucose/metabolism , Humans , Kinetics , Leishmania braziliensis/chemistry , Leishmania braziliensis/genetics , Models, Molecular , Phosphorylation , Protozoan Proteins/genetics , Sequence Alignment , Substrate Specificity
7.
Mol Biochem Parasitol ; 204(2): 64-76, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26778112

ABSTRACT

Glucokinase and hexokinase from pathogenic protozoa Trypanosoma cruzi are potential drug targets for antiparasitic chemotherapy of Chagas' disease. These glucose kinases phosphorylate d-glucose with co-substrate ATP and yield glucose 6-phosphate and are involved in essential metabolic pathways, such as glycolysis and the pentose phosphate pathway. An inhibitor class was conceived that is selective for T. cruzi glucokinase (TcGlcK) using structure-based drug design involving glucosamine having a linker from the C2 amino that terminates with a hydrophobic group either being phenyl, p-hydroxyphenyl, or dioxobenzo[b]thiophenyl groups. The synthesis and characterization for two of the four compounds are presented while the other two compounds were commercially available. Four high-resolution X-ray crystal structures of TcGlcK inhibitor complexes are reported along with enzyme inhibition constants (Ki) for TcGlcK and Homo sapiens hexokinase IV (HsHxKIV). These glucosamine analogue inhibitors include three strongly selective TcGlcK inhibitors and a fourth inhibitor, benzoyl glucosamine (BENZ-GlcN), which is a similar variant exhibiting a shorter linker. Carboxybenzyl glucosamine (CBZ-GlcN) was found to be the strongest glucokinase inhibitor known to date, having a Ki of 0.71±0.05µM. Also reported are two biologically active inhibitors against in vitro T. cruzi culture that were BENZ-GlcN and CBZ-GlcN, with intracellular amastigote growth inhibition IC50 values of 16.08±0.16µM and 48.73±0.69µM, respectively. These compounds revealed little to no toxicity against mammalian NIH-3T3 fibroblasts and provide a key starting point for further drug development with this class of compound.


Subject(s)
Antiprotozoal Agents/chemistry , Enzyme Inhibitors/chemistry , Glucokinase/antagonists & inhibitors , Glucosamine/analogs & derivatives , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , Trypanosoma cruzi/enzymology , Antiprotozoal Agents/metabolism , Chagas Disease/parasitology , Drug Design , Enzyme Inhibitors/metabolism , Glucokinase/chemistry , Glucokinase/genetics , Glucokinase/metabolism , Glucosamine/metabolism , Humans , Kinetics , Models, Molecular , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Trypanosoma cruzi/chemistry , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/genetics
8.
Supramol Chem ; 27(1-2): 65-71, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25506191

ABSTRACT

The (+) and (-) enantiomers for a cryptophane-7-bond-linker-benzenesulfonamide biosensor (C7B) were synthesized and their chirality confirmed by electronic circular dichroism (ECD) spectroscopy. Biosensor binding to carbonic anhydrase II (CAII) was characterized for both enantiomers by hyperpolarized (hp) 129Xe NMR spectroscopy. Our previous study of the racemic (+/-) C7B biosensor-CAII complex [Chambers, et al., J. Am. Chem. Soc. 2009, 131, 563-569], identified two "bound" 129Xe@C7B peaks by hp 129Xe NMR (at 71 and 67 ppm, relative to "free" biosensor at 64 ppm), which led to the initial hypothesis that (+) and (-) enantiomers produce diastereomeric peaks when coordinated to Zn2+ at the chiral CAII active site. Unexpectedly, the single enantiomers complexed with CAII also identified two "bound" 129Xe@C7B peaks: (+) 72, 68 ppm and (-) 68, 67 ppm. These results are consistent with X-ray crystallographic evidence for benzenesulfonamide inhibitors occupying a second site near the CAII surface. As illustrated by our studies of this model protein-ligand interaction, hp 129Xe NMR spectroscopy can be useful for identifying supramolecular assemblies in solution.

9.
Biochemistry ; 52(32): 5441-53, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23905850

ABSTRACT

Aristolochene synthase, a metal-dependent sesquiterpene cyclase from Aspergillus terreus, catalyzes the ionization-dependent cyclization of farnesyl diphosphate (FPP) to form the bicyclic eremophilane (+)-aristolochene with perfect structural and stereochemical precision. Here, we report the X-ray crystal structure of aristolochene synthase complexed with three Mg(2+) ions and the unreactive substrate analogue farnesyl-S-thiolodiphosphate (FSPP), showing that the substrate diphosphate group is anchored by metal coordination and hydrogen bond interactions identical to those previously observed in the complex with three Mg(2+) ions and inorganic pyrophosphate (PPi). Moreover, the binding conformation of FSPP directly mimics that expected for productively bound FPP, with the exception of the precise alignment of the C-S bond with regard to the C10-C11 π system that would be required for C1-C10 bond formation in the first step of catalysis. We also report crystal structures of aristolochene synthase complexed with Mg(2+)3-PPi and ammonium or iminium analogues of bicyclic carbocation intermediates proposed for the natural cyclization cascade. Various binding orientations are observed for these bicyclic analogues, and these orientations appear to be driven by favorable electrostatic interactions between the positively charged ammonium group of the analogue and the negatively charged PPi anion. Surprisingly, the active site is sufficiently flexible to accommodate analogues with partially or completely incorrect stereochemistry. Although this permissiveness in binding is unanticipated, based on the stereochemical precision of catalysis that leads exclusively to the (+)-aristolochene stereoisomer, it suggests the ability of the active site to enable controlled reorientation of intermediates during the cyclization cascade. Taken together, these structures illuminate important aspects of the catalytic mechanism.


Subject(s)
Isomerases/chemistry , Aspergillus/enzymology , Aspergillus/metabolism , Binding Sites , Catalysis , Catalytic Domain , Cations, Divalent , Diphosphates/chemistry , Diphosphates/metabolism , Hydrogen Bonding , Isomerases/metabolism , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Polyisoprenyl Phosphates/chemistry , Polyisoprenyl Phosphates/metabolism , Quaternary Ammonium Compounds/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/metabolism , Stereoisomerism
10.
Arch Biochem Biophys ; 535(2): 163-76, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23583962

ABSTRACT

Arginase from parasitic protozoa belonging to the genus Leishmania is a potential drug target for the treatment of leishmaniasis because this binuclear manganese metalloenzyme catalyzes the first committed step in the biosynthesis of polyamines that enable cell growth and survival. The high resolution X-ray crystal structures of the unliganded form of Leishmania mexicana arginase (LmARG) and four inhibitor complexes are now reported. These complexes include the reactive substrate analogue 2(S)-amino-6-boronohexanoic acid (ABH) and the hydroxylated substrate analogue nor-N(ω)-hydroxy-l-arginine (nor-NOHA), which are the most potent arginase inhibitors known to date. Comparisons of the LmARG structure with that of the archetypal arginase, human arginase I, reveal that all residues important for substrate binding and catalysis are strictly conserved. However, three regions of tertiary structure differ between the parasitic enzyme and the human enzyme corresponding to the G62 - S71, L161 - C172, and I219 - V230 segments of LmARG. Additionally, variations are observed in salt link interactions that stabilize trimer assembly in LmARG. We also report biological studies in which we demonstrate that localization of LmARG to the glycosome, a unique subcellular organelle peculiar to Leishmania and related parasites, is essential for robust pathogenesis.


Subject(s)
Arginase/chemistry , Leishmania mexicana/enzymology , Protozoan Proteins/chemistry , Aminocaproates/chemistry , Aminocaproates/pharmacology , Animals , Antiparasitic Agents/pharmacology , Arginase/antagonists & inhibitors , Arginine/analogs & derivatives , Arginine/chemistry , Arginine/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Crystallography, X-Ray , Female , Humans , Leishmania mexicana/drug effects , Leishmaniasis, Cutaneous/enzymology , Leishmaniasis, Cutaneous/parasitology , Mice , Mice, Inbred BALB C , Models, Molecular , Ornithine/chemistry , Polyamines/antagonists & inhibitors , Polyamines/metabolism , Protein Conformation , Protein Multimerization , Protozoan Proteins/antagonists & inhibitors
11.
Proteins ; 81(6): 1051-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23401043

ABSTRACT

The reversible acetylation of lysine to form N6-acetyllysine in the regulation of protein function is a hallmark of epigenetics. Acetylation of the positively charged amino group of the lysine side chain generates a neutral N-alkylacetamide moiety that serves as a molecular "switch" for the modulation of protein function and protein-protein interactions. We now report the analysis of 381 N6-acetyllysine side chain amide conformations as found in 79 protein crystal structures and 11 protein NMR structures deposited in the Protein Data Bank (PDB) of the Research Collaboratory for Structural Bioinformatics. We find that only 74.3% of N6-acetyllysine residues in protein crystal structures and 46.5% in protein NMR structures contain amide groups with energetically preferred trans or generously trans conformations. Surprisingly, 17.6% of N6-acetyllysine residues in protein crystal structures and 5.3% in protein NMR structures contain amide groups with energetically unfavorable cis or generously cis conformations. Even more surprisingly, 8.1% of N6-acetyllysine residues in protein crystal structures and 48.2% in NMR structures contain amide groups with energetically prohibitive twisted conformations that approach the transition state structure for cis-trans isomerization. In contrast, 109 unique N-alkylacetamide groups contained in 84 highly accurate small molecule crystal structures retrieved from the Cambridge Structural Database exclusively adopt energetically preferred trans conformations. Therefore, we conclude that cis and twisted N6-acetyllysine amides in protein structures deposited in the PDB are erroneously modeled due to their energetically unfavorable or prohibitive conformations.


Subject(s)
Amides/chemistry , Lysine/analogs & derivatives , Proteins/chemistry , Databases, Protein , Isomerism , Lysine/chemistry , Models, Molecular , Molecular Conformation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Thermodynamics
12.
Biochemistry ; 51(42): 8399-409, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23061982

ABSTRACT

Various binuclear metal ion clusters and complexes have been reconstituted in crystalline human arginase I by removing the Mn(2+)(2) cluster of the wild-type enzyme with metal chelators and subsequently soaking the crystalline apoenzyme in buffer solutions containing NiCl(2) or ZnCl(2). X-ray crystal structures of these metal ion variants are correlated with catalytic activity measurements that reveal differences resulting from metal ion substitution. Additionally, treatment of crystalline Mn(2+)(2)-human arginase I with Zn(2+) reveals for the first time the structural basis for inhibition by Zn(2+), which forms a carboxylate-histidine-Zn(2+) triad with H141 and E277. The imidazole side chain of H141 is known to be hyper-reactive, and its chemical modification or mutagenesis is known to similarly compromise catalysis. The reactive substrate analogue 2(S)-amino-6-boronohexanoic acid (ABH) binds as a tetrahedral boronate anion to Mn(2+)(2), Co(2+)(2), Ni(2+)(2), and Zn(2+)(2) clusters in human arginase I, and it can be stabilized by a third inhibitory Zn(2+) ion coordinated by H141. Because ABH binds as an analogue of the tetrahedral intermediate and its flanking transition states in catalysis, this implies that the various metallo-substituted enzymes are capable of some level of catalysis with an actual substrate. Accordingly, we establish the following trend for turnover number (k(cat)) and catalytic efficiency (k(cat)/K(M)): Mn(2+) > Ni(2+) ≈ Co(2+) ≫ Zn(2+). Therefore, Mn(2+) is required for optimal catalysis by human arginase I.


Subject(s)
Arginase/chemistry , Aminocaproates/chemistry , Aminocaproates/metabolism , Arginase/antagonists & inhibitors , Arginase/metabolism , Boron Compounds/chemistry , Boron Compounds/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Manganese/chemistry , Models, Molecular , Nickel/chemistry , Zinc/chemistry
13.
Article in English | MEDLINE | ID: mdl-22869115

ABSTRACT

Human arginase I (HAI) is a binuclear manganese metalloenzyme that catalyzes the hydrolysis of L-arginine to form L-ornithine and urea through a metal-activated hydroxide mechanism. Since HAI regulates L-Arg bioavailability for NO biosynthesis, it is a potential drug target for the treatment of cardiovascular diseases such as atherosclerosis. X-ray crystal structures are now reported of the complexes of Mn(2)(2+)-HAI and Co(2)(2+)-HAI with L-2-amino-3-guanidinopropionic acid (AGPA; also known as dinor-L-arginine), an amino acid bearing a guanidinium side chain two methylene groups shorter than that of L-arginine. Hydrogen bonds to the α-carboxylate and α-amino groups of AGPA dominate enzyme-inhibitor recognition; the guanidinium group does not interact directly with the metal ions.


Subject(s)
Alanine/analogs & derivatives , Arginase/chemistry , Alanine/chemistry , Alanine/metabolism , Arginase/metabolism , Biocatalysis , Humans , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Substrate Specificity
14.
Biochemistry ; 50(44): 9664-80, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-21950839

ABSTRACT

The proximal side of dehaloperoxidase-hemoglobin A (DHP A) from Amphitrite ornata has been modified via site-directed mutagenesis of methionine 86 into aspartate (M86D) to introduce an Asp-His-Fe triad charge relay. X-ray crystallographic structure determination of the metcyano forms of M86D [Protein Data Bank (PDB) entry 3MYN ] and M86E (PDB entry 3MYM ) mutants reveal the structural origins of a stable catalytic triad in DHP A. A decrease in the rate of H(2)O(2) activation as well as a lowered reduction potential versus that of the wild-type enzyme was observed in M86D. One possible explanation for the significantly lower activity is an increased affinity for the distal histidine in binding to the heme Fe to form a bis-histidine adduct. Resonance Raman spectroscopy demonstrates a pH-dependent ligation by the distal histidine in M86D, which is indicative of an increased trans effect. At pH 5.0, the heme Fe is five-coordinate, and this structure resembles the wild-type DHP A resting state. However, at pH 7.0, the distal histidine appears to form a six-coordinate ferric bis-histidine (hemichrome) adduct. These observations can be explained by the effect of the increased positive charge on the heme Fe on the formation of a six-coordinate low-spin adduct, which inhibits the ligation and activation of H(2)O(2) as required for peroxidase activity. The results suggest that the proximal charge relay in peroxidases regulate the redox potential of the heme Fe but that the trans effect is a carefully balanced property that can both activate H(2)O(2) and attract ligation by the distal histidine. To understand the balance of forces that modulate peroxidase reactivity, we studied three M86 mutants, M86A, M86D, and M86E, by spectroelectrochemistry and nuclear magnetic resonance spectroscopy of (13)C- and (15)N-labeled cyanide adducts as probes of the redox potential and of the trans effect in the heme Fe, both of which can be correlated with the proximity of negative charge to the N(δ) hydrogen of the proximal histidine, consistent with an Asp-His-Fe charge relay observed in heme peroxidases.


Subject(s)
Aspartic Acid/chemistry , Catalytic Domain , Globins/chemistry , Histidine/chemistry , Animals , Aspartic Acid/genetics , Catalytic Domain/genetics , Crystallography, X-Ray , Electrochemistry , Globins/genetics , Helminth Proteins/chemistry , Helminth Proteins/genetics , Histidine/genetics , Magnetic Resonance Spectroscopy , Mutagenesis, Site-Directed , Polychaeta/enzymology , Polychaeta/genetics , Spectrophotometry, Ultraviolet , Spectrum Analysis, Raman
15.
Biochemistry ; 50(37): 8018-27, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21870783

ABSTRACT

The binuclear manganese metalloenzyme human arginase I (HAI) is a potential protein drug for cancer chemotherapy, in that it is capable of depleting extracellular l-Arg levels in the microenvironment of tumor cells that require this nutrient to thrive. Substitution of the native Mn(2+)(2) cluster with a Co(2+)(2) cluster in the active site yields an enzyme with enhanced catalytic activity at physiological pH (∼7.4) that could serve as an improved protein drug for L-Arg depletion therapy [Stone, E. M., Glazer, E. S., Chantranupong, L., Cherukuri, P., Breece, R. M., Tierney, D. L., Curley, S. A., Iverson, B. L., and Georgiou, G. (2010) ACS Chem. Biol. 5, 333-342]. A different catalytic mechanism is proposed for Co(2+)(2)-HAI compared with that of Mn(2+)(2)-HAI, including an unusual Nε-Co(2+) coordination mode, to rationalize the lower K(M) value of L-Arg and the lower K(i) value of L-Orn. However, we now report that no unusual metal coordination modes are observed in the cobalt-reconstituted enzyme. The X-ray crystal structures of unliganded Co(2+)(2)-HAI determined at 2.10 Šresolution (pH 7.0) and 1.97 Šresolution (pH 8.5), as well as the structures of Co(2+)(2)-HAI complexed with the reactive substrate analogue 2(S)-amino-6-boronohexanoic acid (ABH, pH 7.0) and the catalytic product L-Orn (pH 7.0) determined at 1.85 and 1.50 Šresolution, respectively, are essentially identical to the corresponding structures of Mn(2+)(2)-HAI. Therefore, in the absence of significant structural differences between Co(2+)(2)-HAI and Mn(2+)(2)-HAI, we suggest that a higher concentration of metal-bridging hydroxide ion at physiological pH for Co(2+)(2)-HAI, a consequence of the lower pK(a) of a Co(2+)-bound water molecule compared with a Mn(2+)-bound water molecule, strengthens electrostatic interactions with cationic amino acids and accounts for enhanced affinity as reflected in the lower K(M) value of L-Arg and the lower K(i) value of L-Orn.


Subject(s)
Arginase/biosynthesis , Arginase/chemistry , Cobalt/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Cobalt/metabolism , Crystallography, X-Ray/methods , Humans , Protein Structure, Secondary
16.
Biochemistry ; 49(31): 6600-16, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20545299

ABSTRACT

Dehaloperoxidase (DHP) from the terebellid polychaete Amphitrite ornata is a bifunctional enzyme that possesses both hemoglobin and peroxidase activities. Of the two DHP isoenzymes identified to date, much of the recent focus has been on DHP A, whereas very little is known pertaining to the activity, substrate specificity, mechanism of function, or spectroscopic properties of DHP B. Herein, we report the recombinant expression and purification of DHP B, as well as the details of our investigations into its catalytic cycle using biochemical assays, stopped-flow UV-visible, resonance Raman, and rapid freeze-quench electron paramagnetic resonance spectroscopies, and spectroelectrochemistry. Our experimental design reveals mechanistic insights and kinetic descriptions of the dehaloperoxidase mechanism which have not been previously reported for isoenzyme A. Namely, we demonstrate a novel reaction pathway in which the products of the oxidative dehalogenation of trihalophenols (dihaloquinones) are themselves capable of inducing formation of oxyferrous DHP B, and an updated catalytic cycle for DHP is proposed. We further demonstrate that, unlike the traditional monofunctional peroxidases, the oxyferrous state in DHP is a peroxidase-competent starting species, which suggests that the ferric oxidation state may not be an obligatory starting point for the enzyme. The data presented herein provide a link between the peroxidase and oxygen transport activities which furthers our understanding of how this bifunctional enzyme is able to unite its two inherent functions in one system.


Subject(s)
Peroxidases/metabolism , Polychaeta/enzymology , Animals , Cloning, Molecular , Halogenation , Hemoglobins , Iron , Kinetics , Oxidation-Reduction , Oxygen/metabolism , Peroxidases/genetics , Spectrum Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...