Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 415
Filter
1.
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
2.
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
3.
Nucleic Acids Res ; 49(19): 11038-11049, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34643703

ABSTRACT

A key pathway for mRNA degradation in bacterial cells begins with conversion of the initial 5'-terminal triphosphate to a monophosphate, a modification that renders transcripts more vulnerable to attack by ribonucleases whose affinity for monophosphorylated 5' ends potentiates their catalytic efficacy. In Escherichia coli, the only proteins known to be important for controlling degradation via this pathway are the RNA pyrophosphohydrolase RppH, its heteromeric partner DapF, and the 5'-monophosphate-assisted endonucleases RNase E and RNase G. We have now identified the metabolic enzyme cytidylate kinase as another protein that affects rates of 5'-end-dependent mRNA degradation in E. coli. It does so by utilizing two distinct mechanisms to influence the 5'-terminal phosphorylation state of RNA, each dependent on the catalytic activity of cytidylate kinase and not its mere presence in cells. First, this enzyme acts in conjunction with DapF to stimulate the conversion of 5' triphosphates to monophosphates by RppH. In addition, it suppresses the direct synthesis of monophosphorylated transcripts that begin with cytidine by reducing the cellular concentration of cytidine monophosphate, thereby disfavoring the 5'-terminal incorporation of this nucleotide by RNA polymerase during transcription initiation. Together, these findings suggest dual signaling pathways by which nucleotide metabolism can impact mRNA degradation in bacteria.


Subject(s)
Cytidine Monophosphate/metabolism , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Phosphotransferases/genetics , RNA Stability/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Nucleoside-Phosphate Kinase/genetics , Nucleoside-Phosphate Kinase/metabolism , Phosphorylation , Phosphotransferases/metabolism , RNA, Bacterial/genetics , RNA, Messenger/genetics , Signal Transduction
4.
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
5.
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
6.
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
7.
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
8.
Mol Biol Rep ; 47(5): 3719-3733, 2020 May.
Article in English | MEDLINE | ID: mdl-32338332

ABSTRACT

Streptococcus mutans UA159 is responsible for human dental caries with robust cariogenic potential. Our previous study noted that a glutamate racemase (MurI) mutant strain (designated S. mutans FW1718), with the hereditary background of UA159, displayed alterations of morphogenesis, attenuated stress tolerance, and weakened biofilm-forming capabilities, accompanying with unclear mechanisms. In this study, we applied isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics to characterize the proteome profiles of the murI mutant strain vs. the wild-type strain in chemically defined media to elucidate the mechanisms by which S. mutans copes with MurI deficiency. Whole-cell proteins of S. mutans FW1718 and UA159 were assessed by iTRAQ-coupled LC-ESI-MS/MS. Furthermore, differentially expressed proteins (DEPs) were identified by Mascot, Gene Ontology (GO) annotation, Cluster of Orthologous Groups of proteins (COG), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Finally, a protein-protein interaction (PPI) network was established using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). Among 1173 total bacterial proteins identified, 112 DEPs exhibited altered expression patterns in S. mutans UA159 with or without the murI mutation. The ΔmurI cells displayed an increase in the relative expression of 93 proteins (fold change ≥ 1.2, p < 0.05) and a decrease in 29 proteins (fold change ≤ 0.833, p < 0.05) compared with the wild-type cells. PPI analysis revealed a complex network of DEPs containing 191 edges and 122 nodes. The DEPs significantly upregulated after murI knockout had roles in diverse functional processes spanning cell-wall biosynthesis, energy production, and DNA replication and repair. We identified distinct variations and diverse modulators caused by murI mutation in the proteome of S. mutans, indicating that the modification of cell membrane structure, redistribution of energy metabolism and enhanced nucleic acid machinery contributed to the S. mutans response to specific environmental contexts.


Subject(s)
Amino Acid Isomerases/metabolism , Streptococcus mutans/metabolism , Amino Acid Isomerases/genetics , Bacterial Proteins/genetics , Biofilms/growth & development , Culture Media/chemistry , Dental Caries/microbiology , Gene Expression Regulation, Bacterial/genetics , Gene Ontology , Protein Interaction Maps/genetics , Proteome/metabolism , Proteomics/methods , Streptococcus mutans/genetics , Tandem Mass Spectrometry/methods
9.
Biochemistry (Mosc) ; 85(2): 248-256, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32093601

ABSTRACT

Streptococcus iniae is a pathogenic and zoonotic bacterium responsible for human diseases and mortality of many fish species. Recently, this bacterium has demonstrated an increasing trend for antibiotics resistance, which has warranted a search for new approaches to tackle its infection. Glutamate racemase (MurI) is a ubiquitous enzyme of the peptidoglycan synthesis pathway that plays an important role in the cell wall integrity maintenance; however, the significance of this enzyme differs in different species. In this study, we knocked out the MurI gene in S. iniae in order to elucidate the role of glutamate racemase in maintaining cell wall integrity in this bacterial species. We also cloned, expressed, and purified MurI and determined its biochemical characteristics. Biochemical analysis revealed that the MurI gene in S. iniae encodes a functional enzyme with a molecular weight of 30 kDa, temperature optimum at 35°C, and pH optimum at 8.5. Metal ions, such as Cu2+, Mn2+, Co2+ and Zn2+, inhibited the enzyme activity. MurI was found to be essential for the viability and cell wall integrity of S. iniae. The optimal growth of the MurI-deficient S. iniae mutant can be achieved only by adding a high concentration of D-glutamate to the medium. Membrane permeability assay of the mutant showed an increasing extent of the cell wall damage with time upon D-glutamate starvation. Moreover, the mutant lost its virulence when incubated in fish blood. Our results demonstrated that the MurI knockout leads to the generation of S. iniae auxotroph with damaged cell walls.


Subject(s)
Amino Acid Isomerases/metabolism , Cell Wall , Microbial Viability , Streptococcus iniae/enzymology , Amino Acid Isomerases/antagonists & inhibitors , Amino Acid Isomerases/genetics , Cell Wall/drug effects , Cell Wall/metabolism , Hydrogen-Ion Concentration , Metals, Heavy/pharmacology , Microbial Viability/drug effects , Mutation , Streptococcus iniae/drug effects , Streptococcus iniae/metabolism
10.
Mol Microbiol ; 107(2): 198-213, 2018 01.
Article in English | MEDLINE | ID: mdl-29134701

ABSTRACT

Glutamate racemase (MurI) has been proposed as a target for anti-tuberculosis drug development based on the inability of ΔmurI mutants of Mycobacterium smegmatis to grow in the absence of d-glutamate. In this communication, we identify ΔmurI suppressor mutants that are detected during prolonged incubation. Whole genome sequencing of these ΔmurI suppressor mutants identified the presence of a SNP, located in the promoter region of MSMEG_5795. RT-qPCR and transcriptional fusion analyses revealed that the ΔmurI suppressor mutant overexpressed MSMEG_5795 14-fold compared to the isogenic wild-type. MSMEG_5795, which is annotated as 4-amino-4-deoxychorismate lyase (ADCL) but which also has homology to d-amino acid transaminase (d-AAT), was expressed, purified and found to have d-AAT activity and to be capable of producing d-glutamate from d-alanine. Consistent with its d-amino acid transaminase function, overexpressed MSMEG_5795 is able to complement both ΔmurI deletion mutants and alanine racemase (Δalr) deletion mutants, thus confirming a multifunctional role for this enzyme in M. smegmatis.


Subject(s)
Amino Acid Isomerases/metabolism , D-Alanine Transaminase/metabolism , Mycobacterium smegmatis/enzymology , Oxo-Acid-Lyases/metabolism , Alanine/metabolism , Alanine Racemase/genetics , Alanine Racemase/metabolism , Amino Acid Isomerases/genetics , Base Sequence/genetics , D-Alanine Transaminase/chemistry , D-Alanine Transaminase/genetics , Gene Deletion , Glutamic Acid/metabolism , Mycobacterium smegmatis/genetics , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/genetics , Promoter Regions, Genetic , Suppression, Genetic , Whole Genome Sequencing
11.
Anal Chem ; 91(17): 11355-11361, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31359753

ABSTRACT

Time-of-flight secondary ion mass spectrometry (ToF-SIMS) using a (CO2)6k+ gas cluster ion beam (GCIB) was used to analyze Escherichia coli mutants previously identified as having impaired plasmid transfer capability related to the spread of antibiotic resistance. The subset of mutants selected were expected to result in changes in the bacterial envelope composition through the deletion of genes encoding for FabF, DapF, and Lpp, where the surface sensitivity of ToF-SIMS can be most useful. Analysis of arrays of spotted bacteria allowed changes in the lipid composition of the bacteria to be elucidated using multivariate analysis and confirmed through imaging of individual ion signals. Significant changes in chemical composition were observed, including a surprising loss of cyclopropanated fatty acids in the fabF mutant where FabF is associated with the elongation of FA(16:1) to FA(18:1) and not cyclopropane formation. The ability of the GCIB to generate increased higher mass signals from biological samples allowed intact lipid A (m/z 1796) to be detected on the bacteria and, despite a 40 keV impact energy, depth profiled through the bacterial envelope along with other high mass ions including species at m/z 1820 and 2428, attributed to ECACYC, that were only observed below the surface of the bacteria and were notably absent in the depth profile of the lpp mutant. The analysis provides new insights into the action of the specific pathways targeted in this study and paves the way for whole new avenues for the characterization of intact molecules within the bacterial envelope.


Subject(s)
Cell Membrane/chemistry , Escherichia coli/genetics , Plasmids/metabolism , Spectrometry, Mass, Secondary Ion/methods , Acetyltransferases/genetics , Amino Acid Isomerases/genetics , Bacterial Outer Membrane Proteins/genetics , Drug Resistance, Microbial , Escherichia coli/chemistry , Escherichia coli/enzymology , Escherichia coli/ultrastructure , Escherichia coli Proteins/genetics , Fatty Acid Synthase, Type II/genetics , Fatty Acids/analysis , Lipid A/analysis , Lipids/analysis , Lipoproteins/genetics , Mutant Proteins
12.
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
13.
J Bacteriol ; 200(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29440254

ABSTRACT

The RidA protein (PF01042) from Salmonella enterica is a deaminase that quenches 2-aminoacrylate (2AA) and other reactive metabolites. In the absence of RidA, 2AA accumulates, damages cellular enzymes, and compromises the metabolic network. In vitro, RidA homologs from all domains of life deaminate 2AA, and RidA proteins from plants, bacteria, yeast, and humans complement the mutant phenotype of a ridA mutant strain of S. enterica In the present study, a methanogenic archaeon, Methanococcus maripaludis S2, was used to probe alternative mechanisms to restore metabolic balance. M. maripaludis MMP0739, which is annotated as an aspartate/glutamate racemase, complemented a ridA mutant strain and reduced the intracellular 2AA burden. The aspartate/glutamate racemase YgeA from Escherichia coli or S. enterica, when provided in trans, similarly restored wild-type growth to a ridA mutant. These results uncovered a new mechanism to ameliorate metabolic stress, and they suggest that direct quenching by RidA is not the only strategy to quench 2AA.IMPORTANCE 2-Aminoacrylate is an endogenously generated reactive metabolite that can damage cellular enzymes if not directly quenched by the conserved deaminase RidA. This study used an archaeon to identify a RidA-independent mechanism to prevent metabolic stress caused by 2AA. The data suggest that a gene product annotated as an aspartate/glutamate racemase (MMP0739) produces a metabolite that can quench 2AA, expanding our understanding of strategies available to quench reactive metabolites.


Subject(s)
Acrylates/chemistry , Bacterial Proteins/metabolism , Pyridoxal Phosphate/metabolism , Racemases and Epimerases/metabolism , Salmonella enterica/genetics , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , Aminohydrolases/genetics , Aminohydrolases/metabolism , Aspartic Acid/metabolism , Bacterial Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Methanococcus/genetics , Methanococcus/metabolism , Racemases and Epimerases/genetics , Salmonella enterica/enzymology
14.
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
15.
Proc Natl Acad Sci U S A ; 112(37): 11726-31, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26324904

ABSTRACT

Plants produce diverse low-molecular-weight compounds via specialized metabolism. Discovery of the pathways underlying production of these metabolites is an important challenge for harnessing the huge chemical diversity and catalytic potential in the plant kingdom for human uses, but this effort is often encumbered by the necessity to initially identify compounds of interest or purify a catalyst involved in their synthesis. As an alternative approach, we have performed untargeted metabolite profiling and genome-wide association analysis on 440 natural accessions of Arabidopsis thaliana. This approach allowed us to establish genetic linkages between metabolites and genes. Investigation of one of the metabolite-gene associations led to the identification of N-malonyl-D-allo-isoleucine, and the discovery of a novel amino acid racemase involved in its biosynthesis. This finding provides, to our knowledge, the first functional characterization of a eukaryotic member of a large and widely conserved phenazine biosynthesis protein PhzF-like protein family. Unlike most of known eukaryotic amino acid racemases, the newly discovered enzyme does not require pyridoxal 5'-phosphate for its activity. This study thus identifies a new d-amino acid racemase gene family and advances our knowledge of plant d-amino acid metabolism that is currently largely unexplored. It also demonstrates that exploitation of natural metabolic variation by integrating metabolomics with genome-wide association is a powerful approach for functional genomics study of specialized metabolism.


Subject(s)
Amino Acid Isomerases/physiology , Arabidopsis Proteins/physiology , Arabidopsis/enzymology , Arabidopsis/genetics , Amino Acid Isomerases/genetics , Arabidopsis Proteins/genetics , Chromatography, High Pressure Liquid , Chromatography, Liquid , Chromosome Mapping , Gene Expression Regulation, Plant , Genetic Variation , Genome-Wide Association Study , Genomics , Genotype , Isoleucine/analogs & derivatives , Isoleucine/chemistry , Mass Spectrometry , Metabolomics , Mutation , Quantitative Trait Loci , Stereoisomerism
16.
Amino Acids ; 49(10): 1743-1754, 2017 10.
Article in English | MEDLINE | ID: mdl-28744579

ABSTRACT

Recently, we cloned and characterized eleven serine and aspartate racemases (SerR and AspR, respectively) from animals. These SerRs and AspRs are not separated by their racemase functions and form a serine/aspartate racemase family cluster based on phylogenetic analysis. Moreover, we have proposed that the AspR-specific triple serine loop region at amino acid positions 150-152 may be responsible for the large AspR activity. In the present study, to test this hypothesis, we prepared and characterized fourteen mutants in this region of animal SerRs and AspRs. The large AspR activity in Acropora and Crassostrea AspR was reduced to <0.04% of wild-type after substitution of the triple serine loop region. Conversely, introducing the triple serine loop region into Acropora, Crassostrea, and Penaeus SerR drastically increased the AspR activity. Those mutants showed similar or higher substrate affinity for aspartate than serine and showed 11-683-fold higher k cat and 28-351-fold higher k cat/K m values for aspartate than serine racemization. Furthermore, we introduced serine residues in all combinations at position 150-152 in mouse SerR. These mutants revealed that a change in the enzyme function from SerR to AspR can be caused by introduction of Ser151 and Ser152, and addition of the third serine residue at position 150 further enhances the enzyme specificity for aspartate due to a decrease in the serine racemase and serine dehydratase activity. Here, we provide convincing evidence that the AspR gene has evolved from the SerR gene by acquisition of the triple serine loop region.


Subject(s)
Amino Acid Isomerases , Anthozoa , Arthropod Proteins , Crassostrea , Mutation, Missense , Penaeidae , Racemases and Epimerases , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Amino Acid Substitution , Animals , Anthozoa/enzymology , Anthozoa/genetics , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Crassostrea/enzymology , Crassostrea/genetics , Mice , Penaeidae/enzymology , Penaeidae/genetics , Protein Structure, Secondary , Racemases and Epimerases/chemistry , Racemases and Epimerases/genetics
17.
Biochem J ; 473(9): 1267-80, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26964898

ABSTRACT

Glutamate racemase (MurI) is responsible for providing D-glutamate for peptidoglycan biosynthesis in bacteria and has been a favoured target in pharmaceutical drug design efforts. It has recently been proven to be essential in Mycobacterium tuberculosis, the causative organism of tuberculosis, a disease for which new medications are urgently needed. In the present study, we have determined the protein crystal structures of MurI from both M. tuberculosis and Mycobacterium smegmatis in complex with D-glutamate to 2.3 Å and 1.8 Å resolution respectively. These structures are conserved, but reveal differences in their active site architecture compared with that of other MurI structures. Furthermore, compounds designed to target other glutamate racemases have been screened but do not inhibit mycobacterial MurI, suggesting that a new drug design effort will be needed to develop inhibitors. A new type of MurI dimer arrangement has been observed in both structures, and this arrangement becomes the third biological dimer geometry for MurI found to date. The mycobacterial MurI dimer is tightly associated, with a KD in the nanomolar range. The enzyme binds D- and L-glutamate specifically, but is inactive in solution unless the dimer interface is mutated. We created triple mutants of this interface in the M. smegmatis glutamate racemase (D26R/R105A/G194R or E) that have appreciable activity (kcat=0.056-0.160 min(-1) and KM=0.26-0.51 mM) and can be utilized to screen proposed antimicrobial candidates for inhibition.


Subject(s)
Amino Acid Isomerases/chemistry , Bacterial Proteins/chemistry , Glutamic Acid/chemistry , Mycobacterium tuberculosis/enzymology , Amino Acid Isomerases/genetics , Amino Acid Substitution , Bacterial Proteins/genetics , Mutation, Missense , Mycobacterium tuberculosis/genetics , Protein Domains
18.
Antimicrob Agents Chemother ; 60(10): 6091-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27480853

ABSTRACT

The increasing global prevalence of drug resistance among many leading human pathogens necessitates both the development of antibiotics with novel mechanisms of action and a better understanding of the physiological activities of preexisting clinically effective drugs. Inhibition of peptidoglycan (PG) biosynthesis and cross-linking has traditionally enjoyed immense success as an antibiotic target in multiple bacterial pathogens, except in Mycobacterium tuberculosis, where it has so far been underexploited. d-Cycloserine, a clinically approved antituberculosis therapeutic, inhibits enzymes within the d-alanine subbranch of the PG-biosynthetic pathway and has been a focus in our laboratory for understanding peptidoglycan biosynthesis inhibition and for drug development in studies of M. tuberculosis During our studies on alternative inhibitors of the d-alanine pathway, we discovered that the canonical alanine racemase (Alr) inhibitor ß-chloro-d-alanine (BCDA) is a very poor inhibitor of recombinant M. tuberculosis Alr, despite having potent antituberculosis activity. Through a combination of enzymology, microbiology, metabolomics, and proteomics, we show here that BCDA does not inhibit the d-alanine pathway in intact cells, consistent with its poor in vitro activity, and that it is instead a mechanism-based inactivator of glutamate racemase (MurI), an upstream enzyme in the same early stage of PG biosynthesis. This is the first report to our knowledge of inhibition of MurI in M. tuberculosis and thus provides a valuable tool for studying this essential and enigmatic enzyme and a starting point for future MurI-targeted antibacterial development.


Subject(s)
Amino Acid Isomerases/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/chemistry , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , beta-Alanine/analogs & derivatives , Amino Acid Isomerases/antagonists & inhibitors , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , Amino Acid Sequence , Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cloning, Molecular , Enzyme Inhibitors/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Microbial Sensitivity Tests , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Peptidoglycan/biosynthesis , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity , beta-Alanine/chemistry , beta-Alanine/pharmacology
19.
Environ Microbiol ; 18(6): 1673-85, 2016 06.
Article in English | MEDLINE | ID: mdl-26419727

ABSTRACT

Enzymes catalysing the stereo-chemical inter-conversion of amino acids are known as amino acid racemases. In bacteria, these enzymes are fundamental to synthesize the D-Ala and D-Glu that are critical components of the peptidoglycan. In addition to this structural function in cell wall assembly, D-amino acids produced by microbial amino acid racemases have been described as relevant constituents in other prokaryotic structures (e.g. capsule, non-ribosomal peptides) and have been associated to growth fitness and to processes such as biofilm development, spore germination and signalling. The recent discovery of broad spectrum racemases able to produce and release several D-amino acids to the environment suggests that these enzymes might have a great impact in microbial ecology. Consequently, new data on the biochemistry and regulation of racemases is key to understand the biological significance of D-enantiomers in nature, in particular their effect on microbial social networks. This review summarizes current knowledge on the environmental roles of bacterial racemases with an emphasis on the potential roles of the new broad spectrum enzymes in natural environments.


Subject(s)
Amino Acid Isomerases/metabolism , Bacteria/enzymology , Bacterial Proteins/metabolism , Environmental Microbiology , Amino Acid Isomerases/genetics , Amino Acids/metabolism , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , Biofilms , Cell Wall/metabolism
20.
Amino Acids ; 48(2): 387-402, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26352274

ABSTRACT

Free D-amino acids have been found in various invertebrate phyla, while amino acid racemase genes have been identified in few species. The purpose of this study is to elucidate the distribution, function, and evolution of amino acid racemases in invertebrate animals. We searched the GenBank databases, and found 11 homologous serine racemase genes from eight species in eight different invertebrate phyla. The cloned genes were identified based on their maximum activity as Acropora millepora (Cnidaria) serine racemase (SerR) and aspartate racemase (AspR), Caenorhabditis elegans (Nematoda) SerR, Capitella teleta (Annelida) SerR, Crassostrea gigas (Mollusca) SerR and AspR, Dugesia japonica (Platyhelminthes) SerR, Milnesium tardigradum (Tardigrada) SerR, Penaeus monodon (Arthropoda) SerR and AspR and Strongylocentrotus purpuratus (Echinodermata) AspR. We found that Acropora, Aplysia, Capitella, Crassostrea and Penaeus had two amino acid racemase paralogous genes and these paralogous genes have evolved independently by gene duplication at their recent ancestral species. The transcriptome analyses using available SRA data and enzyme kinetic data suggested that these paralogous genes are expressed in different tissues and have different functions in vivo. Phylogenetic analyses clearly indicated that animal SerR and AspR are not separated by their particular racemase functions and form a serine/aspartate racemase family cluster. Our results revealed that SerR and AspR are more widely distributed among invertebrates than previously known. Moreover, we propose that the triple serine loop motif at amino acid positions 150-152 may be responsible for the large aspartate racemase activity and the AspR evolution from SerR.


Subject(s)
Amino Acid Isomerases/genetics , Aspartic Acid/metabolism , Invertebrates/enzymology , Racemases and Epimerases/genetics , Serine/metabolism , Amino Acid Isomerases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Escherichia coli/genetics , Invertebrates/genetics , Phylogeny , Pyridoxal Phosphate/metabolism , Racemases and Epimerases/metabolism , Sequence Alignment , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL