Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
1.
J Chem Inf Model ; 64(10): 4250-4262, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38701175

ABSTRACT

The microbial enzyme diaminopimelate epimerase (DapF), a vital enzyme in the lysine biosynthetic pathway, catalyzes the conversion of L, L-diaminopimelate (L, L-DAP) to D, L-diaminopimelate (D, L-DAP) using a catalytic cysteine dyad with one cysteine in thiol state and another in thiolate. Under oxidizing conditions, the catalytic cysteines of apo DapF form a disulfide bond that alters the structure and function of DapF. Given its potential as a target for antimicrobial resistance treatments, understanding DapF's functional dynamics is imperative. In the present work, we employ microsecond-scale all-atom molecular dynamics simulations of product-bound DapF and apo-DapF under oxidized and reduced conditions. We employ a polarized charge model for the ligand and the active site residues, which was necessary to preserve the electrostatic environment in the active site and retain the ligand in the active site. The product-bound DapF and apo-DapF in oxidized and reduced conditions exhibit a closed, semi-open, and open conformation, respectively, as identified using the internal coordinates of the dimeric enzyme and the principal component analysis. The conformational switch is guided by the dynamic catalytic (DC) loop, loop II, and loop III movements in the active site. The time scale of the close-to-open conformational transition is estimated to be 0.8 µs through Markov state modeling (MSM) and transition path theory (TPT). The present study explains the role of various active site residues and loops in ligand binding and protein dynamics in the DapF enzyme under different redox conditions. Such information will be helpful in future inhibitor design studies targeting the DapF enzyme.


Subject(s)
Corynebacterium glutamicum , Markov Chains , Molecular Dynamics Simulation , Protein Conformation , Corynebacterium glutamicum/enzymology , Ligands , Amino Acid Isomerases/metabolism , Amino Acid Isomerases/chemistry , Catalytic Domain , Oxidation-Reduction
2.
Environ Microbiol ; 26(4): e16621, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38558504

ABSTRACT

The Candidate Phyla Radiation (CPR) encompasses widespread uncultivated bacteria with reduced genomes and limited metabolic capacities. Most CPR bacteria lack the minimal set of enzymes required for peptidoglycan (PG) synthesis, leaving it unclear how these bacteria produce this essential envelope component. In this study, we analysed the distribution of d-amino acid racemases that produce the universal PG components d-glutamate (d-Glu) or d-alanine (d-Ala). We also examined moonlighting enzymes that synthesize d-Glu or d-Ala. Unlike other phyla in the domain Bacteria, CPR bacteria do not exhibit these moonlighting activities and have, at most, one gene encoding either a Glu or Ala racemase. One of these 'orphan' racemases is a predicted Glu racemase (MurICPR) from the CPR bacterium Candidatus Saccharimonas aalborgenesis. The expression of MurICPR restores the growth of a Salmonella d-Glu auxotroph lacking its endogenous racemase and results in the substitution of l-Ala by serine as the first residue in a fraction of the PG stem peptides. In vitro, MurICPR exclusively racemizes Glu as a substrate. Therefore, Ca. Saccharimonas aalborgensis may couple Glu racemization to serine and d-Glu incorporation into the stem peptide. Our findings provide the first insights into the synthesis of PG by an uncultivated environmental bacterium and illustrate how to experimentally test enzymatic activities from CPR bacteria related to PG metabolism.


Subject(s)
Amino Acid Isomerases , Amino Acid Isomerases/genetics , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Racemases and Epimerases , Bacteria/metabolism , Glutamic Acid/metabolism , Serine
3.
Plant Cell Environ ; 47(5): 1503-1512, 2024 May.
Article in English | MEDLINE | ID: mdl-38251436

ABSTRACT

d-amino acids are the d stereoisomers of the common l-amino acids found in proteins. Over the past two decades, the occurrence of d-amino acids in plants has been reported and circumstantial evidence for a role in various processes, including interaction with soil microorganisms or interference with cellular signalling, has been provided. However, examples are not numerous and d-amino acids can also be detrimental, some of them inhibiting growth and development. Thus, the persistence of d-amino acid metabolism in plants is rather surprising, and the evolutionary origins of d-amino acid metabolism are currently unclear. Systemic analysis of sequences associated with d-amino acid metabolism enzymes shows that they are not simply inherited from cyanobacterial metabolism. In fact, the history of plant d-amino acid metabolism enzymes likely involves multiple steps, cellular compartments, gene transfers and losses. Regardless of evolutionary steps, enzymes of d-amino acid metabolism, such as d-amino acid transferases or racemases, have been retained by higher plants and have not simply been eliminated, so it is likely that they fulfil important metabolic roles such as serine, folate or plastid peptidoglycan metabolism. We suggest that d-amino acid metabolism may have been critical to support metabolic functions required during the evolution of land plants.


Subject(s)
Amino Acid Isomerases , Embryophyta , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , Amino Acids/metabolism , Plants/metabolism , Embryophyta/metabolism , Bacteria/metabolism
4.
Biochemistry ; 60(50): 3829-3840, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34845903

ABSTRACT

Catalytic promiscuity is the coincidental ability to catalyze nonbiological reactions in the same active site as the native biological reaction. Several lines of evidence show that catalytic promiscuity plays a role in the evolution of new enzyme functions. Thus, studying catalytic promiscuity can help identify structural features that predispose an enzyme to evolve new functions. This study identifies a potentially preadaptive residue in a promiscuous N-succinylamino acid racemase/o-succinylbenzoate synthase (NSAR/OSBS) enzyme from Amycolatopsis sp. T-1-60. This enzyme belongs to a branch of the OSBS family which includes many catalytically promiscuous NSAR/OSBS enzymes. R266 is conserved in all members of the NSAR/OSBS subfamily. However, the homologous position is usually hydrophobic in other OSBS subfamilies, whose enzymes lack NSAR activity. The second-shell amino acid R266 is close to the catalytic acid/base K263, but it does not contact the substrate, suggesting that R266 could affect the catalytic mechanism. Mutating R266 to glutamine in Amycolatopsis NSAR/OSBS profoundly reduces NSAR activity but moderately reduces OSBS activity. This is due to a 1000-fold decrease in the rate of proton exchange between the substrate and the general acid/base catalyst K263. This mutation is less deleterious for the OSBS reaction because K263 forms a cation-π interaction with the OSBS substrate and/or the intermediate, rather than acting as a general acid/base catalyst. Together, the data explain how R266 contributes to NSAR reaction specificity and was likely an essential preadaptation for the evolution of NSAR activity.


Subject(s)
Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Carbon-Carbon Lyases/chemistry , Carbon-Carbon Lyases/metabolism , Amino Acid Isomerases/genetics , Amino Acid Sequence , Amino Acid Substitution , Amycolatopsis/enzymology , Amycolatopsis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Carbon-Carbon Lyases/genetics , Catalytic Domain/genetics , Conserved Sequence , Crystallography, X-Ray , Enzyme Stability/genetics , Evolution, Molecular , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
5.
Biosci Biotechnol Biochem ; 85(7): 1650-1657, 2021 Jun 24.
Article in English | MEDLINE | ID: mdl-33942867

ABSTRACT

The amino acid sequence of the OCC_10945 gene product from the hyperthermophilic archaeon Thermococcus litoralis DSM5473, originally annotated as γ-aminobutyrate aminotransferase, is highly similar to that of the uncharacterized pyridoxal 5'-phosphate (PLP)-dependent amino acid racemase from Pyrococcus horikoshii. The OCC_10945 enzyme was successfully overexpressed in Escherichia coli by coexpression with a chaperone protein. The purified enzyme demonstrated PLP-dependent amino acid racemase activity primarily toward Met and Leu. Although PLP contributed to enzyme stability, it only loosely bound to this enzyme. Enzyme activity was strongly inhibited by several metal ions, including Co2+ and Zn2+, and nonsubstrate amino acids such as l-Arg and l-Lys. These results suggest that the underlying PLP-binding and substrate recognition mechanisms in this enzyme are significantly different from those of the other archaeal and bacterial amino acid racemases. This is the first description of a novel PLP-dependent amino acid racemase with moderate substrate specificity in hyperthermophilic archaea.


Subject(s)
Amino Acid Isomerases/metabolism , Archaeal Proteins/metabolism , Thermococcus/enzymology , Amino Acid Isomerases/chemistry , Amino Acid Sequence , Archaeal Proteins/chemistry , Electrophoresis, Polyacrylamide Gel , Genes, Archaeal , Molecular Chaperones/metabolism , Phylogeny , Substrate Specificity , Thermococcus/genetics
6.
J Mol Recognit ; 34(9): e2894, 2021 09.
Article in English | MEDLINE | ID: mdl-33719110

ABSTRACT

Enterococcus faecalis (E. faecalis) is a Gram-positive coccoid, non-sporulating, facultative anaerobic, multidrug resistance bacterium responsible for almost 65% to 80% of all enterococcal nosocomial infections. It usually causes infective endocarditis, urinary tract and surgical wound infections. The increase in E. faecalis resistance to conventionally available antibiotic has rekindled intense interest in developing useful antibacterial drugs. In E. faecalis, diaminopimelate epimerase (DapF) is involved in the lysine biosynthetic pathway. The product of this pathway is precursors of peptidoglycan synthesis, which is a component of bacterial cell wall. Also, because mammals lack this enzyme, consequently E. faecalis diaminopimelate epimerase (EfDapF) represents a potential target for developing novel class of antibiotics. In this regard, we have successfully cloned, overexpressed the gene encoding DapF in BL-21(DE3) and purified with Ni-NTA Agarose resin. In addition to this, binding studies were performed using fluorescence spectroscopy in order to confirm the bindings of the identified lead compounds (acetaminophen and dexamethasone) with EfDapF. Docking studies revealed that acetaminophen found to make hydrogen bonds with Asn72 and Asn13 while dexamethasone interacted by forming hydrogen bonds with Asn205 and Glu223. Thus, biochemical studies indicated acetaminophen and dexamethasone, as potential inhibitors of EfDapF and eventually can reduce the catalytic activity of EfDapF.


Subject(s)
Acetaminophen/pharmacology , Amino Acid Isomerases/antagonists & inhibitors , Dexamethasone/pharmacology , Enterococcus faecalis/enzymology , Molecular Docking Simulation , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Drug Repositioning , Enterococcus faecalis/drug effects , Protein Conformation
7.
J Bacteriol ; 203(7)2021 03 08.
Article in English | MEDLINE | ID: mdl-33468590

ABSTRACT

Members of Thermococcales harbor a number of genes encoding putative aminotransferase class III enzymes. Here, we characterized the TK1211 protein from the hyperthermophilic archaeon Thermococcus kodakarensis The TK1211 gene was expressed in T. kodakarensis under the control of the strong, constitutive promoter of the cell surface glycoprotein gene TK0895 (P csg ). The purified protein did not display aminotransferase activity but exhibited racemase activity. An examination of most amino acids indicated that the enzyme was a racemase with relatively high activity toward Leu and Met. Kinetic analysis indicated that Leu was the most preferred substrate. A TK1211 gene disruption strain (ΔTK1211) was constructed and grown on minimal medium supplemented with l- or d-Leu or l- or d-Met. The wild-type T. kodakarensis is not able to synthesize Leu and displays Leu auxotrophy, providing a direct means to examine the Leu racemase activity of the TK1211 protein in vivo When we replaced l-Leu with d-Leu in the medium, the host strain with an intact TK1211 gene displayed an extended lag phase but displayed cell yield similar to that observed in medium with l-Leu. In contrast, the ΔTK1211 strain displayed growth in medium with l-Leu but could not grow with d-Leu. The results indicate that TK1211 encodes a Leu racemase that is active in T. kodakarensis cells and that no other protein exhibits this activity, at least to an extent that can support growth. Growth experiments with l- or d-Met also confirmed the Met racemase activity of the TK1211 protein in T. kodakarensisIMPORTANCE Phylogenetic analysis of aminotransferase class III proteins from all domains of life reveals numerous groups of protein sequences. One of these groups includes a large number of sequences from Thermococcales species and can be divided into four subgroups. Representatives of three of these subgroups have been characterized in detail. This study reveals that a representative from the remaining uncharacterized subgroup is an amino acid racemase with preference toward Leu and Met. Taken together with results of previous studies on enzymes from Pyrococcus horikoshii and Thermococcus kodakarensis, members of the four subgroups now can be presumed to function as a broad-substrate-specificity amino acid racemase (subgroup 1), alanine/serine racemase (subgroup 2), ornithine ω-aminotransferase (subgroup 3), or Leu/Met racemase (subgroup 4).


Subject(s)
Amino Acid Isomerases/metabolism , Archaeal Proteins/metabolism , Thermococcus/enzymology , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Amino Acid Sequence , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Hot Temperature , Kinetics , Leucine/metabolism , Methionine/metabolism , Phylogeny , Substrate Specificity , Thermococcus/chemistry , Thermococcus/genetics , Thermococcus/metabolism
8.
Biotechnol Bioeng ; 117(9): 2683-2693, 2020 09.
Article in English | MEDLINE | ID: mdl-32492177

ABSTRACT

Enzymes are industrially applied under increasingly diverse environmental conditions that are dictated by the efforts to optimize overall process efficiency. Engineering the operational stability of biocatalysts to enhance their half-lives under the desired process conditions is a widely applied strategy to reduce costs. Here, we present a simple method to enhance enzyme stability in the presence of monophasic aqueous/organic solvent mixtures based on the concept of strengthening the enzyme's surface hydrogen-bond network by exchanging surface-located amino acid residues for arginine. Suitable residues are identified from sequence comparisons with homologous enzymes from thermophilic organisms and combined using a shuffling approach to obtain an enzyme variant with increased stability in monophasic aqueous/organic solvent mixtures. With this approach, we increase the stability of the broad-spectrum amino acid racemase of Pseudomonas putida DSM 3263 eightfold in mixtures with 40% methanol and sixfold in mixtures with 30% acetonitrile.


Subject(s)
Amino Acid Isomerases , Bacterial Proteins , Protein Engineering/methods , Acetonitriles/chemistry , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enzyme Stability , Escherichia coli/genetics , Mutation/genetics , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Solvents/chemistry
9.
Biochim Biophys Acta Proteins Proteom ; 1868(9): 140461, 2020 09.
Article in English | MEDLINE | ID: mdl-32474108

ABSTRACT

d-Amino acids are physiologically important components of peptidoglycan in the bacterial cell wall, maintaining cell structure and aiding adaptation to environmental changes through peptidoglycan remodelling. Therefore, the biosynthesis of d-amino acids is essential for bacteria to adapt to different environmental conditions. The peptidoglycan of the extremely thermophilic bacterium Thermus thermophilus contains d-alanine (d-Ala) and d-glutamate (d-Glu), but its d-amino acid metabolism remains poorly understood. Here, we investigated the enzyme activity and function of the product of the TTHA1643 gene, which is annotated to be a Glu racemase in the T. thermophilus HB8 genome. Among 21 amino acids tested, TTHA1643 showed highly specific activity toward Glu as the substrate. The catalytic efficiency (kcat/Km) of TTHA1643 toward d- and l-Glu was comparable; however, the kcat value was 18-fold higher for l-Glu than for d-Glu. Temperature and pH profiles showed that the racemase activity of TTHA1643 is high under physiological conditions for T. thermophilus growth. To assess physiological relevance, we constructed a TTHA1643-deficient strain (∆TTHA1643) by replacing the TTHA1643 gene with the thermostable hygromycin resistance gene. Growth of the ∆TTHA1643 strain in synthetic medium without d-Glu was clearly diminished relative to wild type, although the TTHA1643 deletion was not lethal, suggesting that alternative d-Glu biosynthetic pathways may exist. The deterioration in growth was restored by adding d-Glu to the culture medium, showing that d-Glu is required for normal growth of T. thermophilus. Collectively, our findings show that TTHA1643 is a Glu racemase and has the physiological function of d-Glu production in T. thermophilus.


Subject(s)
Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , Thermus thermophilus/enzymology , Amino Acid Sequence , Amino Acids/metabolism , Cell Wall/chemistry , Cloning, Molecular , Enzyme Stability , Escherichia coli/metabolism , Gene Deletion , Genome, Bacterial , Glutamic Acid/metabolism , Hydrogen-Ion Concentration , Kinetics , Recombinant Proteins , Substrate Specificity , Temperature , Thermus thermophilus/genetics , Thermus thermophilus/growth & development , Thermus thermophilus/physiology , Transcriptome
10.
Proteins ; 88(11): 1513-1527, 2020 11.
Article in English | MEDLINE | ID: mdl-32543729

ABSTRACT

Protein domains exist by themselves or in combination with other domains to form complex multidomain proteins. Defining domain boundaries in proteins is essential for understanding their evolution and function but is not trivial. More specifically, partitioning domains that interact by forming a single ß-sheet is known to be particularly troublesome for automatic structure-based domain decomposition pipelines. Here, we study edge-to-edge ß-strand interactions between domains in a protein chain, to help define the boundaries for some more difficult cases where a single ß-sheet spanning over two domains gives an appearance of one. We give a number of examples where ß-strands belonging to a single ß-sheet do not belong to a single domain and highlight the difficulties of automatic domain parsers on these examples. This work can be used as a baseline for defining domain boundaries in homologous proteins or proteins with similar domain interactions in the future.


Subject(s)
Amino Acid Isomerases/chemistry , Penicillin-Binding Proteins/chemistry , Protein Interaction Domains and Motifs , Racemases and Epimerases/chemistry , Amino Acid Isomerases/metabolism , Amino Acid Sequence , Animals , Bacteria/chemistry , Binding Sites , Databases, Protein , Datasets as Topic , Humans , Models, Molecular , Penicillin-Binding Proteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Racemases and Epimerases/metabolism , Thermodynamics
11.
Biochim Biophys Acta Proteins Proteom ; 1868(4): 140377, 2020 04.
Article in English | MEDLINE | ID: mdl-31982578

ABSTRACT

The N-succinylamino acid racemase/o-succinylbenzoate synthase (NSAR/OSBS) subfamily from the enolase superfamily contains different enzymes showing promiscuous N-substituted-amino acid racemase (NxAR) activity. These enzymes were originally named as N-acylamino acid racemases because of their industrial application. Nonetheless, they are pivotal in several enzymatic cascades due to their versatility to catalyze a wide substrate spectrum, allowing the production of optically pure d- or l-amino acids from cheap precursors. These compounds are of paramount economic interest, since they are used as food additives, in the pharmaceutical and cosmetics industries and/or as chiral synthons in organic synthesis. Despite its economic importance, the discovery of new N-succinylamino acid racemases has become elusive, since classical sequence-based annotation methods proved ineffective in their identification, due to a high sequence similarity among the members of the enolase superfamily. During the last decade, deeper investigations into different members of the NSAR/OSBS subfamily have shed light on the classification and identification of NSAR enzymes with NxAR activity of biotechnological potential. This review aims to gather the dispersed information on NSAR/OSBS members showing NxAR activity over recent decades, focusing on their biotechnological applications and providing practical advice to identify new enzymes.


Subject(s)
Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Biotechnology , Amino Acid Isomerases/classification , Amino Acid Isomerases/genetics , Biological Evolution , Enzymes, Immobilized , Models, Molecular , Phylogeny , Protein Engineering , Sequence Alignment
12.
Metab Eng ; 59: 15-23, 2020 05.
Article in English | MEDLINE | ID: mdl-31926305

ABSTRACT

Scoring changes in enzyme or pathway performance by their effect on growth behavior is a widely applied strategy for identifying improved biocatalysts. While in directed evolution this strategy is powerful in removing non-functional catalysts in selections, measuring subtle differences in growth behavior remains difficult at high throughput, as it is difficult to focus metabolic control on only one or a few enzymatic steps over the entire process of growth-based discrimination. Here, we demonstrate successful miniaturization of a growth-based directed enzyme evolution process. For cultivation of library clones we employed optically clear gel-like microcarriers of nanoliter volume (NLRs) as reaction vessels and used fluorescence-assisted particle sorting to estimate the growth behavior of each of the gel-embedded clones in a highly parallelized fashion. We demonstrate that the growth behavior correlates with the desired improvements in enzyme performance and that we can fine-tune selection stringency by including an antimetabolite in the assay. As a model enzyme reaction, we improve the racemization of ornithine, a possible starting block for the large-scale synthesis of sulphostin, by a broad-spectrum amino acid racemase and confirm the discriminatory power by showing that even moderately improved enzyme variants can be readily identified.


Subject(s)
Amino Acid Isomerases , Antimetabolites , Directed Molecular Evolution , Organophosphorus Compounds , Piperidones , Protein Engineering , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Antimetabolites/chemical synthesis , Antimetabolites/chemistry , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/chemistry , Piperidones/chemical synthesis , Piperidones/chemistry
13.
Nat Prod Rep ; 36(12): 1687-1705, 2019 12 11.
Article in English | MEDLINE | ID: mdl-30994146

ABSTRACT

Covering: up to March 2019 Amino acid racemases and epimerases are key enzymes that invert the configuration of common amino acids and supply many corresponding d-isomers in living organisms. Some d-amino acids are inherently bioactive, whereas others are building blocks for important biomolecules, for example lipid II, the bacterial cell wall precursor. Peptides containing them have enhanced proteolytic stability and can act as important recognition elements in mammalian systems. Selective inhibition of certain amino acid racemases (e.g. glutamate racemase) is believed to offer a promising target for new antibacterial drugs effective against pathogens resistant to current antibiotics. Many amino acid racemases employ imine formation with pyridoxal phosphate (PLP) as a cofactor to accelerate the abstraction of the alpha proton. However, the group reviewed herein achieves racemization of free amino acids without the use of cofactors or metals, and uses a thiol/thiolate pair for deprotonation and reprotonation. All bacteria and higher plants contain such enzymes, for example diaminopimelate epimerase, which is required for lysine biosynthesis in these organisms. This process cannot be accomplished without an enzyme catalyst as the acidities of a thiol and the substrate α-hydrogen are inherently mismatched by at least 10 orders of magnitude. This review describes the structural and mechanistic studies on PLP-independent racemases and the evolving view of key enzymatic machinery that accomplishes these remarkable transformations.


Subject(s)
Enzyme Inhibitors/pharmacology , Pyridoxal Phosphate/metabolism , Racemases and Epimerases/chemistry , Racemases and Epimerases/metabolism , Amino Acid Isomerases/antagonists & inhibitors , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Enzyme Inhibitors/chemistry , Protein Conformation , Racemases and Epimerases/antagonists & inhibitors , Sulfhydryl Compounds/metabolism
14.
Biochem Biophys Res Commun ; 511(1): 135-140, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30773259

ABSTRACT

The hypothetical OCC_00372 protein from Thermococcus litoralis is a member of the ProR superfamily from hyperthermophilic archaea and exhibits unique bifunctional proline racemase/hydroxyproline 2-epimerase activity. However, the molecular mechanism of the broad substrate specificity and extreme thermostability of this enzyme (TlProR) remains unclear. Here we determined the crystal structure of TlProR at 2.7 Šresolution. Of note, a substrate proline molecule, derived from expression host Escherichia coli cells, was tightly bound in the active site of TlProR. The substrate bound structure and mutational analyses suggested that Trp241 is involved in hydroxyproline recognition by making a hydrogen bond between the indole group of Trp241 and the hydroxyl group of hydroxyproline. Additionally, Tyr171 may contribute to the thermostability by making hydrogen bonds between the hydroxyl group of Tyr171 and catalytic residues. Our structural and functional analyses provide a structural basis for understanding the molecular mechanism of substrate specificity and thermostability of ProR superfamily proteins.


Subject(s)
Amino Acid Isomerases/chemistry , Thermococcus/enzymology , Amino Acid Isomerases/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Enzyme Stability , Hydroxyproline/metabolism , Models, Molecular , Protein Conformation , Substrate Specificity , Thermococcus/chemistry , Thermococcus/metabolism
15.
Amino Acids ; 51(2): 331-343, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30377839

ABSTRACT

We expressed a histidine racemase from Leuconostoc mesenteroides subsp. sake NBRC 102480 (Lm-HisR) successively in a soluble fraction of Escherichia coli BL21 (DE3) and then highly purified it from the cell-free extract. Lm-HisR showed amino acid racemase activity on histidine specifically. This is the first example of an amino acid racemase specifically acting on histidine. Phylogenetic analysis of Lm-HisR showed that Lm-HisR was located far from the cluster of alanine racemases reported thus far and only in lactic acid bacteria of the genus Leuconostoc. Alignment of the primary structure of Lm-HisR with those of lysine and alanine racemases and alanine racemase homologs previously reported revealed that the PLP-binding lysine and catalytic tyrosine were completely conserved, and some residues that are unique to the phylogenetic branch of Lm-HisR, Phe44, Ser45, Thr174, Thr206, His286, Ser287, Phe292, Gly312, Val357, and Ala358 were identified. We determined the crystal structure of Lm-HisR complexed with PLP at a 2.1-Å resolution. The crystal structure contained four molecules (two dimers) in the asymmetric unit. When comparing the 3D structure of Lm-HisR with those of racemases from Geobacillus stearothermophilus and Oenococcus oeni, Met315 was completely conserved, but Val357 was not. In addition, two significant differences were observed between Lm-HisR and G. stearothermophilus alanine racemase. Phe44 and His286 in Lm-HisR corresponded to Tyr43 and Tyr284 in G. stearothermophilus alanine racemase, respectively. Based on the structural analysis, comparison with alanine racemase, and docking simulation, three significant residues, Phe44, His286, and Val357, were identified that may control the substrate specificity of Lm-HisR.


Subject(s)
Amino Acid Isomerases/chemistry , Amino Acid Isomerases/isolation & purification , Histidine/chemistry , Leuconostoc mesenteroides/enzymology , Alanine Racemase/chemistry , Amino Acid Isomerases/genetics , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/enzymology , Geobacillus stearothermophilus/enzymology , Hydrogen Bonding , Models, Molecular , Molecular Docking Simulation , Phylogeny , Protein Structure, Secondary , Pyridoxal Phosphate/chemistry
16.
Appl Microbiol Biotechnol ; 103(2): 843-851, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30456576

ABSTRACT

Increasing drug resistance in pathogens including Mycobacterium tuberculosis (MTB) has been ascribed to mutations in the known target genes. However, many of these drugs have multiple targets; some of which have not been identified so far. Understanding the mechanism of action of these drugs holds a great promise in better management of disease especially by drug-resistant strains. In this study, we report glutamate racemase (MurI), a crucial enzyme of phase I peptidoglycan (PG) biosynthesis pathway of MTB, as an additional target of ethambutol (EMB). The effect on EMB on the MurI protein at structural and functional level was studied using different spectroscopic, biochemical, and insilico approaches. Spectroscopic analysis revealed that EMB-modified protein undergoes conformational alterations. Furthermore, in vitro racemization studies of the MurI protein suggest that EMB decreases its functional activity. Docking studies revealed that EMB interacts with most of the active residues at the binding site and blocks the binding pocket. Overall, data suggests that EMB, a primary drug used for the treatment of tuberculosis (TB), acts as a competitive inhibitor of substrate for binding to mycobacterial MurI protein. The study also points out to our lacunae in understanding the site and mechanism of action of existing drugs. Furthermore, glutamate racemase is a conserved protein of the bacterial kingdom; therefore, ethambutol could be a promising candidate as a broad-spectrum antibiotic for many other bacterial diseases.


Subject(s)
Amino Acid Isomerases/antagonists & inhibitors , Antitubercular Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ethambutol/pharmacology , Mycobacterium tuberculosis/drug effects , Peptidoglycan/biosynthesis , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Binding Sites , Cell Wall/metabolism , Molecular Docking Simulation , Mycobacterium tuberculosis/enzymology , Protein Binding , Protein Conformation , Spectrum Analysis
17.
PLoS Negl Trop Dis ; 12(10): e0006853, 2018 10.
Article in English | MEDLINE | ID: mdl-30372428

ABSTRACT

Chagas disease, caused by Trypanosoma cruzi, affects millions of people in South America and no satisfactory therapy exists, especially for its life threatening chronic phase. We targeted the Proline Racemase of T. cruzi, which is present in all stages of the parasite life cycle, to discover new inhibitors against this disease. The first published crystal structures of the enzyme revealed that the catalytic site is too small to allow any relevant drug design. In previous work, to break through the chemical space afforded to virtual screening and drug design, we generated intermediate models between the open (ligand free) and closed (ligand bound) forms of the enzyme. In the present work, we co-crystallized the enzyme with the selected inhibitors and found that they were covalently bound to the catalytic cysteine residues in the active site, thus explaining why these compounds act as irreversible inhibitors. These results led us to the design of a novel, more potent specific inhibitor, NG-P27. Co-crystallization of this new inhibitor with the enzyme allowed us to confirm the predicted protein functional motions and further characterize the chemical mechanism. Hence, the catalytic Cys300 sulfur atom of the enzyme attacks the C2 carbon of the inhibitor in a coupled, regiospecific-stereospecific Michael reaction with trans-addition of a proton on the C3 carbon. Strikingly, the six different conformations of the catalytic site in the crystal structures reported in this work had key similarities to our intermediate models previously generated by inference of the protein functional motions. These crystal structures span a conformational interval covering roughly the first quarter of the opening mechanism, demonstrating the relevance of modeling approaches to break through chemical space in drug design.


Subject(s)
Amino Acid Isomerases/antagonists & inhibitors , Amino Acid Isomerases/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Trypanosoma cruzi/enzymology , Catalytic Domain , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Protein Binding , Protein Conformation
18.
FEBS Lett ; 592(20): 3399-3413, 2018 10.
Article in English | MEDLINE | ID: mdl-30194685

ABSTRACT

Glutamate racemases (GR) catalyze the racemization of d- and l-glutamate and are targets for the development of antibiotics. We demonstrate that GR from the periodontal pathogen Fusobacterium nucleatum (FnGR) catalyzes the racemization of d-homocysteic acid (d-HCA), while l-HCA is a poor substrate. This enantioselectivity arises because l-HCA perturbs FnGR's monomer-dimer equilibrium toward inactive monomer. The inhibitory effect of l-HCA may be overcome by increasing the total FnGR concentration or by adding glutamate, but not by blocking access to the active site through site-directed mutagenesis, suggesting that l-HCA binds at an allosteric site. This phenomenon is also exhibited by GR from Bacillus subtilis, suggesting that enantiospecific, "substrate"-induced dissociation of oligomers to form inactive monomers may furnish a new inhibition strategy.


Subject(s)
Amino Acid Isomerases/chemistry , Bacterial Proteins/chemistry , Homocysteine/analogs & derivatives , Protein Structure, Quaternary , Allosteric Site , Amino Acid Isomerases/metabolism , Bacillus subtilis/enzymology , Bacterial Proteins/metabolism , Biocatalysis , Catalytic Domain , Fusobacterium nucleatum/enzymology , Homocysteine/chemistry , Homocysteine/metabolism , Kinetics , Stereoisomerism , Substrate Specificity
19.
Nucleic Acids Res ; 46(13): 6880-6892, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29931175

ABSTRACT

mRNA decay is an important strategy by which bacteria can rapidly adapt to their ever-changing surroundings. The 5'-terminus state of mRNA determines the velocity of decay of many types of RNA. In Escherichia coli, RNA pyrophosphohydrolase (RppH) is responsible for the removal of the 5'-terminal triphosphate from hundreds of mRNAs and triggers its rapid degradation by ribonucleases. A diaminopimelate epimerase, DapF, can directly interact with RppH and stimulate its hydrolysis activity in vivo and in vitro. However, the molecular mechanism remains to be elucidated. Here, we determined the complex structure of DapF-RppH as a heterotetramer in a 2:2 molar ratio. DapF-bound RppH exhibits an RNA-favorable conformation similar to the RNA-bound state, suggesting that association with DapF promotes and stabilizes RppH in a conformation that facilitates substrate RNA binding and thus stimulates the activity of RppH. To our knowledge, this is the first published structure of an RNA-pyrophosphohydrolysis complex in bacteria. Our study provides a framework for further investigation of the potential regulators involved in the RNA-pyrophosphohydrolysis process in prokaryotes.


Subject(s)
Acid Anhydride Hydrolases/chemistry , Amino Acid Isomerases/chemistry , Escherichia coli Proteins/chemistry , Acid Anhydride Hydrolases/metabolism , Amino Acid Isomerases/metabolism , Crystallography, X-Ray , Escherichia coli Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , RNA/chemistry , RNA/metabolism
20.
Biochemistry ; 57(26): 3676-3689, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29767960

ABSTRACT

Studying the evolution of catalytically promiscuous enzymes like those from the N-succinylamino acid racemase/ o-succinylbenzoate synthase (NSAR/OSBS) subfamily can reveal mechanisms by which new functions evolve. Some enzymes in this subfamily have only OSBS activity, while others catalyze OSBS and NSAR reactions. We characterized several NSAR/OSBS subfamily enzymes as a step toward determining the structural basis for evolving NSAR activity. Three enzymes were promiscuous, like most other characterized NSAR/OSBS subfamily enzymes. However, Alicyclobacillus acidocaldarius OSBS (AaOSBS) efficiently catalyzes OSBS activity but lacks detectable NSAR activity. Competitive inhibition and molecular modeling show that AaOSBS binds N-succinylphenylglycine with moderate affinity in a site that overlaps its normal substrate. On the basis of possible steric conflicts identified by molecular modeling and sequence conservation within the NSAR/OSBS subfamily, we identified one mutation, Y299I, that increased NSAR activity from undetectable to 1.2 × 102 M-1 s-1 without affecting OSBS activity. This mutation does not appear to affect binding affinity but instead affects kcat, by reorienting the substrate or modifying conformational changes to allow both catalytic lysines to access the proton that is moved during the reaction. This is the first site known to affect reaction specificity in the NSAR/OSBS subfamily. However, this gain of activity was obliterated by a second mutation, M18F. Epistatic interference by M18F was unexpected because a phenylalanine at this position is important in another NSAR/OSBS enzyme. Together, modest NSAR activity of Y299I AaOSBS and epistasis between sites 18 and 299 indicate that additional sites influenced the evolution of NSAR reaction specificity in the NSAR/OSBS subfamily.


Subject(s)
Alicyclobacillus/enzymology , Amino Acid Isomerases/metabolism , Carbon-Carbon Lyases/metabolism , Alicyclobacillus/chemistry , Alicyclobacillus/genetics , Alicyclobacillus/metabolism , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Carbon-Carbon Lyases/chemistry , Carbon-Carbon Lyases/genetics , Catalytic Domain , Crystallography, X-Ray , Evolution, Molecular , Models, Molecular , Phylogeny , Protein Conformation , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...