Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 7936, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29786696

ABSTRACT

Dihydrodipicolinate reductase (DHDPR) is a key enzyme in the diaminopimelate- and lysine-synthesis pathways that reduces DHDP to tetrahydrodipicolinate. Although DHDPR uses both NADPH and NADH as a cofactor, the structural basis for cofactor specificity and preference remains unclear. Here, we report that Paenisporosarcina sp. TG-14 PaDHDPR has a strong preference for NADPH over NADH, as determined by isothermal titration calorimetry and enzymatic activity assays. We determined the crystal structures of PaDHDPR alone, with its competitive inhibitor (dipicolinate), and the ternary complex of the enzyme with dipicolinate and NADPH, with results showing that only the ternary complex had a fully closed conformation and suggesting that binding of both substrate and nucleotide cofactor is required for enzymatic activity. Moreover, NADPH binding induced local conformational changes in the N-terminal long loop (residues 34-59) of PaDHDPR, as the His35 and Lys36 residues in this loop interacted with the 2'-phosphate group of NADPH, possibly accounting for the strong preference of PaDHDPR for NADPH. Mutation of these residues revealed reduced NADPH binding and enzymatic activity, confirming their importance in NADPH binding. These findings provide insight into the mechanism of action and cofactor selectivity of this important bacterial enzyme.


Subject(s)
Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/metabolism , NADP/metabolism , Planococcaceae/enzymology , Amino Acid Sequence , Crystallography, X-Ray , Kinetics , Models, Molecular , NADP/chemistry , Protein Conformation , Sequence Homology , Substrate Specificity
2.
Biotechnol Bioeng ; 115(7): 1764-1777, 2018 07.
Article in English | MEDLINE | ID: mdl-29574741

ABSTRACT

l-lysine is an important amino acid in animals and humans and NADPH is a vital cofactor for maximizing the efficiency of l-lysine fermentation. Dihydrodipicolinate reductase (DHDPR), an NAD(P)H-dependent enzyme, shows a variance in nucleotide-cofactor affinity in bacteria. In this study, we rationally engineered Corynebacterium glutamicum DHDPR (CgDHDPR) to switch its nucleotide-cofactor specificity resulting in an increase in final titer (from 82.6 to 117.3 g L-1 ), carbon yield (from 0.35 to 0.44 g [g glucose]-1 ) and productivity (from 2.07 to 2.93 g L-1 hr-1 ) of l-lysine in JL-6 ΔdapB::Ec-dapBC115G,G116C in fed-batch fermentation. To do this, we comparatively analyzed the characteristics of CgDHDPR and Escherichia coli DHDPR (EcDHDPR), indicating that hetero-expression of NADH-dependent EcDHDPR increased l-lysine production. Subsequently, we rationally modified the conserved structure of cofactor-binding motif, and results indicated that introducing the mutation K11A or R13A in CgDHDPR and introducing the mutation R16A or R39A in EcDHDPR modifies the nucleotide-cofactor affinity of DHDPR. Lastly, the effects of these mutated DHDPRs on l-lysine production were investigated. The highest increase (26.2%) in l-lysine production was observed for JL-6 ΔdapB::Ec-dapBC115G,G116C , followed by JL-6 Cg-dapBC37G,G38C (21.4%) and JL-6 ΔdapB::Ec-dapBC46G,G47C (15.2%). This is the first report of a rational modification of DHDPR that enhances the l-lysine production and yield through the modulation of nucleotide-cofactor specificity.


Subject(s)
Coenzymes/metabolism , Corynebacterium glutamicum/enzymology , Dihydrodipicolinate Reductase/genetics , Dihydrodipicolinate Reductase/metabolism , Lysine/metabolism , Nucleotides/metabolism , Amino Acid Substitution , Escherichia coli/genetics , Escherichia coli/metabolism , Fermentation , Mutant Proteins/genetics , Mutant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
Biochem J ; 475(1): 137-150, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29187521

ABSTRACT

Dihydrodipicolinate reductase (DHDPR) catalyses the second reaction in the diaminopimelate pathway of lysine biosynthesis in bacteria and plants. In contrast with the tetrameric bacterial DHDPR enzymes, we show that DHDPR from Vitis vinifera (grape) and Selaginella moellendorffii are dimeric in solution. In the present study, we have also determined the crystal structures of DHDPR enzymes from the plants Arabidopsis thaliana and S. moellendorffii, which are the first dimeric DHDPR structures. The analysis of these models demonstrates that the dimer forms through the intra-strand interface, and that unique secondary features in the plant enzymes block tetramer assembly. In addition, we have also solved the structure of tetrameric DHDPR from the pathogenic bacteria Neisseria meningitidis Measuring the activity of plant DHDPR enzymes showed that they are much more prone to substrate inhibition than the bacterial enzymes, which appears to be a consequence of increased flexibility of the substrate-binding loop and higher affinity for the nucleotide substrate. This higher propensity to substrate inhibition may have consequences for ongoing efforts to increase lysine biosynthesis in plants.


Subject(s)
Bacterial Proteins/chemistry , Dihydrodipicolinate Reductase/chemistry , Picolinic Acids/chemistry , Plant Proteins/chemistry , Vitis/enzymology , Amino Acid Motifs , Arabidopsis/chemistry , Arabidopsis/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , Dihydrodipicolinate Reductase/genetics , Dihydrodipicolinate Reductase/metabolism , Gene Expression , Kinetics , Lysine/biosynthesis , Models, Molecular , NAD/chemistry , NAD/metabolism , NADP/chemistry , NADP/metabolism , Neisseria meningitidis/chemistry , Neisseria meningitidis/enzymology , Picolinic Acids/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Selaginellaceae/chemistry , Selaginellaceae/enzymology , Species Specificity , Substrate Specificity , Vitis/chemistry
4.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 12): 885-891, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27917836

ABSTRACT

In bacteria, the second committed step in the diaminopimelate/lysine anabolic pathways is catalyzed by the enzyme dihydrodipicolinate reductase (DapB). DapB catalyzes the reduction of dihydrodipicolinate to yield tetrahydrodipicolinate. Here, the cloning, expression, purification, crystallization and X-ray diffraction analysis of DapB from the human-pathogenic bacterium Bartonella henselae, the causative bacterium of cat-scratch disease, are reported. Protein crystals were grown in conditions consisting of 5%(w/v) PEG 4000, 200 mM sodium acetate, 100 mM sodium citrate tribasic pH 5.5 and were shown to diffract to ∼2.3 Šresolution. They belonged to space group P4322, with unit-cell parameters a = 109.38, b = 109.38, c = 176.95 Å. Rr.i.m. was 0.11, Rwork was 0.177 and Rfree was 0.208. The three-dimensional structural features of the enzymes show that DapB from B. henselae is a tetramer consisting of four identical polypeptides. In addition, the substrate NADP+ was found to be bound to one monomer, which resulted in a closed conformational change in the N-terminal domain.


Subject(s)
Bacterial Proteins/chemistry , Bartonella henselae/chemistry , Dihydrodipicolinate Reductase/chemistry , NADP/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bartonella henselae/enzymology , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Dihydrodipicolinate Reductase/genetics , Dihydrodipicolinate Reductase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Models, Molecular , NADP/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
5.
PLoS One ; 11(1): e0146525, 2016.
Article in English | MEDLINE | ID: mdl-26815040

ABSTRACT

The enzyme dihydrodipicolinate reductase (DHDPR) is a component of the lysine biosynthetic pathway in bacteria and higher plants. DHDPR catalyzes the NAD(P)H dependent reduction of 2,3-dihydrodipicolinate to the cyclic imine L-2,3,4,5,-tetrahydropicolinic acid. The dapB gene that encodes dihydrodipicolinate reductase has previously been cloned, but the expression of the enzyme is low and the purification is time consuming. Therefore the E. coli dapB gene was cloned into the pET16b vector to improve the protein expression and simplify the purification. The dapB gene sequence was utilized to design forward and reverse oligonucleotide primers that were used to PCR the gene from Escherichia coli genomic DNA. The primers were designed with NdeI or BamHI restriction sites on the 5'and 3' terminus respectively. The PCR product was sequenced to confirm the identity of dapB. The gene was cloned into the expression vector pET16b through NdeI and BamHI restriction endonuclease sites. The resulting plasmid containing dapB was transformed into the bacterial strain BL21 (DE3). The transformed cells were utilized to grow and express the histidine-tagged reductase and the protein was purified using Ni-NTA affinity chromatography. SDS/PAGE gel analysis has shown that the protein was 95% pure and has approximate subunit molecular weight of 28 kDa. The protein purification is completed in one day and 3 liters of culture produced approximately 40-50 mgs of protein, an improvement on the previous protein expression and multistep purification.


Subject(s)
Dihydrodipicolinate Reductase/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Recombinant Fusion Proteins/biosynthesis , Chromatography, Affinity , Cloning, Molecular , Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Histidine/genetics , Histidine/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism , Plasmids/genetics , Plasmids/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification
6.
J Microbiol Biotechnol ; 26(2): 226-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26502738

ABSTRACT

Dihydrodipicolinate reductase is an enzyme that converts dihydrodipicolinate to tetrahydrodipicolinate using an NAD(P)H cofactor in L-lysine biosynthesis. To increase the understanding of the molecular mechanisms of lysine biosynthesis, we determined the crystal structure of dihydrodipicolinate reductase from Corynebacterium glutamicum (CgDapB). CgDapB functions as a tetramer, and each protomer is composed of two domains, an Nterminal domain and a C-terminal domain. The N-terminal domain mainly contributes to nucleotide binding, whereas the C-terminal domain is involved in substrate binding. We elucidated the mode of cofactor binding to CgDapB by determining the crystal structure of the enzyme in complex with NADP(+) and found that CgDapB utilizes both NADH and NADPH as cofactors. Moreover, we determined the substrate binding mode of the enzyme based on the coordination mode of two sulfate ions in our structure. Compared with Mycobacterium tuberculosis DapB in complex with its cofactor and inhibitor, we propose that the domain movement for active site constitution occurs when both cofactor and substrate bind to the enzyme.


Subject(s)
Corynebacterium glutamicum/enzymology , Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/metabolism , Lysine/biosynthesis , NADP/metabolism , NAD/metabolism , Amino Acid Sequence , Binding Sites , Biocatalysis , Corynebacterium glutamicum/metabolism , Crystallography, X-Ray , Kinetics , Models, Molecular , Mycobacterium tuberculosis/enzymology , Oxidoreductases/metabolism , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity
7.
PLoS One ; 10(3): e0118861, 2015.
Article in English | MEDLINE | ID: mdl-25756623

ABSTRACT

Arabidopsis CRINKLY4 (ACR4) is a receptor-like kinase (RLK) involved in the global development of the plant. The Arabidopsis genome encodes four homologs of ACR4 that contain sequence similarity and analogous architectural elements to ACR4, termed Arabidopsis CRINKLY4 Related (AtCRRs) proteins. Additionally, a signaling module has been previously proposed including a postulated peptide ligand, CLE40, the ACR4 RLK, and the WOX5 transcription factor that engage in a possible feedback mechanism controlling stem cell differentiation. However, little biochemical evidence is available to ascertain the molecular aspects of receptor heterodimerization and the role of phosphorylation in these interactions. Therefore, we have undertaken an investigation of the in vitro interactions between the intracellular domains (ICD) of ACR4, the CRRs and WOX5. We demonstrate that interaction can occur between ACR4 and all four CRRs in the unphosphorylated state. However, phosphorylation dependency is observed for the interaction between ACR4 and CRR3. Furthermore, sequence analysis of the ACR4 gene family has revealed a conserved 'KDSAF' motif that may be involved in protein-protein interactions among the receptor family. We demonstrate that peptides harboring this conserved motif in CRR3 and CRK1are able to bind to the ACR4 kinase domain. Our investigations also indicate that the ACR4 ICD can interact with and phosphorylate the transcription factor WOX5.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Arabidopsis/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Binding Sites , Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
8.
Protein Expr Purif ; 85(1): 66-76, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22776412

ABSTRACT

Given the rise of multi drug resistant bacterial strains, such as methicillin-resistant Staphylococcus aureus (MRSA), there is an urgent need to discover new antimicrobial agents. A validated but as yet unexplored target for new antibiotics is dihydrodipicolinate reductase (DHDPR), an enzyme that catalyzes the second step of the lysine biosynthesis pathway in bacteria. We report here the cloning, expression and purification of N-terminally his-tagged recombinant DHDPR from MRSA (6H-MRSA-DHDPR) and compare its secondary and quaternary structure with the wild type (MRSA-DHDPR) enzyme. Comparative analyses demonstrate that recombinant 6H-MRSA-DHDPR is folded and adopts the native tetrameric quaternary structure in solution. Furthermore, kinetic studies show 6H-MRSA-DHDPR is functional, displaying parameters for K(m)(NADH) of 6.0 µM, K(m)(DHDP) of 22 µM, and k(cat) of 21s(-1), which are similar to those reported for the native enzyme. The solution properties and stability of the 6H-MRSA-DHDPR enzyme are also reported in varying physicochemical conditions.


Subject(s)
Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/metabolism , Methicillin-Resistant Staphylococcus aureus/enzymology , Cloning, Molecular , Dihydrodipicolinate Reductase/genetics , Dihydrodipicolinate Reductase/isolation & purification , Enzyme Stability , Histidine/chemistry , Histidine/genetics , Histidine/isolation & purification , Histidine/metabolism , Kinetics , Methicillin-Resistant Staphylococcus aureus/chemistry , Methicillin-Resistant Staphylococcus aureus/genetics , Osmolar Concentration , Protein Conformation , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
9.
FEBS Lett ; 585(16): 2561-7, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21803042

ABSTRACT

Lysine biosynthesis proceeds by the nucleotide-dependent reduction of dihydrodipicolinate (DHDP) to tetrahydrodipicolinate (THDP) by dihydrodipicolinate reductase (DHDPR). The S. aureus DHDPR structure reveals different conformational states of this enzyme even in the absence of a substrate or nucleotide-cofactor. Despite lacking a conserved basic residue essential for NADPH interaction, S. aureus DHDPR differs from other homologues as NADPH is a more preferred co-factor than NADH. The structure provides a rationale-Lys35 compensates for the co-factor site mutation. These observations are significant for bi-ligand inhibitor design that relies on ligand-induced conformational changes as well as co-factor specificity for this important drug target.


Subject(s)
Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/metabolism , NADP/metabolism , Staphylococcus aureus/enzymology , Amino Acid Sequence , Crystallography, X-Ray , Dihydrodipicolinate Reductase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Substrate Specificity
10.
Biochim Biophys Acta ; 1814(12): 1900-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21803176

ABSTRACT

To gain insights into the role of quaternary structure in the TIM-barrel family of enzymes, we introduced mutations to the DHDPS enzyme of Thermotoga maritima, which we have previously shown to be a stable tetramer in solution. These mutations were aimed at reducing the number of salt bridges at one of the two tetramerization interface of the enzyme, which contains many more interactions than the well characterized equivalent interface of the mesophilic Escherichia coli DHDPS enzyme. The resulting variants had altered quaternary structure, as shown by analytical ultracentrifugation, gel filtration liquid chromatography, and small angle X-ray scattering, and X-ray crystallographic studies confirmed that one variant existed as an independent monomer, but with few changes to the secondary and tertiary structure. Reduction of higher order assembly resulted in a loss of thermal stability, as measured by a variety of methods, and impaired catalytic function. Binding of pyruvate increased the oligomeric status of the variants, with a concomitant increase in thermal stability, suggesting a role for substrate binding in optimizing stable, higher order structures. The results of this work show that the salt bridges located at the tetramerization interface of DHDPS play a significant role in maintaining higher order structures, and demonstrate the importance of quaternary structure in determining protein stability and in the optimization of enzyme catalysis.


Subject(s)
Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/metabolism , Protein Multimerization/physiology , Calibration , Cloning, Molecular , Dihydrodipicolinate Reductase/genetics , Dihydrodipicolinate Reductase/isolation & purification , Genetic Variation , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/isolation & purification , Mutant Proteins/metabolism , Protein Binding/genetics , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Multimerization/genetics , Protein Structure, Quaternary , Protein Structure, Secondary , Substrate Specificity/genetics , Thermotoga maritima/chemistry , Thermotoga maritima/enzymology , Thermotoga maritima/genetics , Thermotoga maritima/metabolism
11.
Arch Biochem Biophys ; 512(2): 167-74, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21704017

ABSTRACT

Given the rapid rise in antibiotic resistance, including methicillin resistance in Staphylococcus aureus (MRSA), there is an urgent need to characterize novel drug targets. Enzymes of the lysine biosynthesis pathway in bacteria are examples of such targets, including dihydrodipicolinate reductase (DHDPR, E.C. 1.3.1.26), which is the product of an essential bacterial gene. DHDPR catalyzes the NAD(P)H-dependent reduction of dihydrodipicolinate (DHDP) to tetrahydrodipicolinate (THDP) in the lysine biosynthesis pathway. We show that MRSA-DHDPR exhibits a unique nucleotide specificity utilizing NADPH (K(m)=12µM) as a cofactor more effectively than NADH (K(m)=26µM). However, the enzyme is inhibited by high concentrations of DHDP when using NADPH as a cofactor, but not with NADH. Isothermal titration calorimetry (ITC) studies reveal that MRSA-DHDPR has ∼20-fold greater binding affinity for NADPH (K(d)=1.5µM) relative to NADH (K(d)=29µM). Kinetic investigations in tandem with ITC studies show that the enzyme follows a compulsory-order ternary complex mechanism; with inhibition by DHDP through the formation of a nonproductive ternary complex with NADP(+). This work describes, for the first time, the catalytic mechanism and cofactor preference of MRSA-DHDPR, and provides insight into rational approaches to inhibiting this valid antimicrobial target.


Subject(s)
Dihydrodipicolinate Reductase/metabolism , Methicillin-Resistant Staphylococcus aureus/enzymology , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calorimetry , Catalysis , Dihydrodipicolinate Reductase/antagonists & inhibitors , Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Methicillin-Resistant Staphylococcus aureus/genetics , Molecular Sequence Data , NAD/metabolism , NADP/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Thermodynamics
12.
Metab Eng ; 12(4): 341-51, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20381632

ABSTRACT

In the present work the Gram-positive bacterium Corynebacterium glutamicum was engineered into an efficient, tailor-made production strain for diaminopentane (cadaverine), a highly attractive building block for bio-based polyamides. The engineering comprised expression of lysine decarboxylase (ldcC) from Escherichia coli, catalyzing the conversion of lysine into diaminopentane, and systems-wide metabolic engineering of central supporting pathways. Substantially re-designing the metabolism yielded superior strains with desirable properties such as (i) the release from unwanted feedback regulation at the level of aspartokinase and pyruvate carboxylase by introducing the point mutations lysC311 and pycA458, (ii) an optimized supply of the key precursor oxaloacetate by amplifying the anaplerotic enzyme, pyruvate carboxylase, and deleting phosphoenolpyruvate carboxykinase which otherwise removes oxaloacetate, (iii) enhanced biosynthetic flux via combined amplification of aspartokinase, dihydrodipicolinate reductase, diaminopimelate dehydrogenase and diaminopimelate decarboxylase, and (iv) attenuated flux into the threonine pathway competing with production by the leaky mutation hom59 in the homoserine dehydrogenase gene. Lysine decarboxylase proved to be a bottleneck for efficient production, since its in vitro activity and in vivo flux were closely correlated. To achieve an optimal strain having only stable genomic modifications, the combination of the strong constitutive C. glutamicum tuf promoter and optimized codon usage allowed efficient genome-based ldcC expression and resulted in a high diaminopentane yield of 200 mmol mol(-1). By supplementing the medium with 1 mgL(-1) pyridoxal, the cofactor of lysine decarboxylase, the yield was increased to 300 mmol mol(-1). In the production strain obtained, lysine secretion was almost completely abolished. Metabolic analysis, however, revealed substantial formation of an as yet unknown by-product. It was identified as an acetylated variant, N-acetyl-diaminopentane, which reached levels of more than 25% of that of the desired product.


Subject(s)
Cadaverine/biosynthesis , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Metabolic Networks and Pathways/genetics , Amino Acid Oxidoreductases/metabolism , Aspartate Kinase/genetics , Aspartate Kinase/metabolism , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Dihydrodipicolinate Reductase/metabolism , Feedback, Physiological , Gene Expression Regulation, Bacterial , Genetic Engineering , Homoserine Dehydrogenase/genetics , Homoserine Dehydrogenase/metabolism , Lysine/metabolism , Oxaloacetic Acid/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Point Mutation , Pyridoxal/metabolism , Pyruvate Carboxylase/genetics , Pyruvate Carboxylase/metabolism , Systems Biology , Threonine/metabolism
13.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 1): 61-72, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20057050

ABSTRACT

Dihydrodipicolinate reductase (DHDPR, DapB) is an enzyme that belongs to the L-lysine biosynthetic pathway. DHDPR reduces the alpha,beta-unsaturated cyclic imine 2,3-dihydrodipicolinic acid to yield the compound 2,3,4,5-tetrahydrodipicolinic acid in a pyridine nucleotide-dependent reaction. The substrate of this reaction is the unstable product of the preceding enzyme dihydrodipicolinate synthase (DHDPS, DapA). Here, the structure of apo-DHDPR from Mycobacterium tuberculosis is reported in two orthorhombic crystal forms, as well as the structure of DHDPR from M. tuberculosis in complex with NADH in a monoclinic crystal form. A comparison of the results with previously solved structures of this enzyme shows that DHDPR undergoes a major conformational change upon binding of its cofactor. This conformational change can be interpreted as one of the low-frequency normal modes of the structure.


Subject(s)
Bacterial Proteins/chemistry , Dihydrodipicolinate Reductase/chemistry , Mycobacterium tuberculosis/enzymology , NAD/chemistry , Allosteric Regulation , Bacterial Proteins/metabolism , Crystallization , Crystallography, X-Ray , Dihydrodipicolinate Reductase/metabolism , Hydro-Lyases/metabolism , NAD/metabolism , Protein Binding , Protein Conformation , Pyridines/metabolism
14.
Plant Cell ; 21(3): 928-43, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19318609

ABSTRACT

Inducible high-affinity copper uptake is key to copper homeostasis in Chlamydomonas reinhardtii. We generated cDNAs and updated gene models for four genes, CTR1, CTR2, CTR3, and COPT1, encoding CTR-type copper transporters in Chlamydomonas. The expression of CTR1, CTR2, and CTR3 increases in copper deficient cells and in response to hypoxia or Ni(2+) supplementation; this response depends on the transcriptional activator CRR1. A copper response element was identified by mutational analysis of the 5' upstream region of CTR1. Functional analyses identify CTR1 and CTR2 as the assimilatory transporters of Chlamydomonas based on localization to the plasma membrane and ability to rescue a Saccharomyces cerevisiae mutant defective in high-affinity copper transport. The Chlamydomonas CTRs contain a novel Cys-Met motif (CxxMxxMxxC-x(5/6)-C), which occurs also in homologous proteins in other green algae, amoebae, and pathogenic fungi. CTR3 appears to have arisen by duplication of CTR2, but CTR3 lacks the characteristic transmembrane domains found in the transporters, suggesting that it may be a soluble protein. Thus, Chlamydomonas CTR genes encode a distinct subset of the classical CTR family of Cu(I) transporters and represent new targets of CRR1-dependent signaling.


Subject(s)
Amino Acid Motifs , Cation Transport Proteins/metabolism , Cell Membrane/metabolism , Chlamydomonas reinhardtii/metabolism , Copper/metabolism , Protein Isoforms/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cation Transport Proteins/genetics , Chlamydomonas reinhardtii/cytology , Chlamydomonas reinhardtii/genetics , Copper Transporter 1 , Dihydrodipicolinate Reductase/genetics , Dihydrodipicolinate Reductase/metabolism , Gene Expression Regulation , Genes, Reporter , Homeostasis , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Nickel/chemistry , Protein Isoforms/genetics , Protozoan Proteins/genetics , Sequence Alignment
15.
Biochemistry ; 47(38): 9966-80, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18710263

ABSTRACT

Dihydrodipicolinate reductase (DHPR) is a homotetramer that catalyzes reduction of dihydrodipicolinate (DHP). We recently reported a biligand inhibitor ( K i = 100 nM) of DHPR, comprised of fragments that bind in the NADH (CRAA = catechol rhodanine acetic acid) and DHP (PDC = pyridine dicarboxylate) binding sites. Herein, we characterize binding synergy and cooperativity for ligand binding to Escherichia coli DHPR: NADH or CRAA and PDC (stable analog of DHP). While K d values indicate little synergy between NADH and PDC, (1)H- (15)N HSQC chemical shift perturbation and saturation transfer difference (STD) titrations indicate that PDC induces a more dramatic conformational change than NADH, consistent with a role in domain closure. PDC binds cooperatively (Hill coefficient = 2), while NADH does not, based on STD titrations that monitor only fast exchange processes. However, HSQC titrations monitoring Trp253 (located between monomers) indicate that NADH binds in two steps, with high affinity binding to only one of the monomers. Therefore, DHPR binds cofactor via a sequential model, with negative cooperativity. These results, interpreted in light of steady-state data, suggest that DHPR activity requires NADH binding at only one of the four monomers. Implications of our results for fragment assembly are discussed, using CRAA tethering to PDC as a model biligand: (a) if one fragment (ex. PDC) must induce a large structural change before the other fragment is brought proximal, this must be screened for upfront, and (b) cooperative or synergistic interactions between binding sites can lead to unexpected and misleading effects in NMR-based screening.


Subject(s)
Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/metabolism , Enzyme Inhibitors/chemical synthesis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Binding Sites/physiology , Dihydrodipicolinate Reductase/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Ligands , Substrate Specificity/physiology
16.
J Biochem ; 143(5): 617-23, 2008 May.
Article in English | MEDLINE | ID: mdl-18250105

ABSTRACT

In lysine biosynthesis, dihydrodipicolinate reductase (DHDPR) catalyses the formation of tetrahydrodipicolinate. Unlike DHDPR enzymes from Escherichia coli and Mycobacterium tuberculosis, which have dual specificity for both NADH and NADPH as co-factors, the enzyme from Thermotoga maritima has a significantly greater affinity for NADPH. Despite low sequence identity with the E. coli and M. tuberculosis DHDPR enzymes, DHDPR from T. maritima has a similar catalytic site, with many conserved residues involved in interactions with substrates. This suggests that as the enzyme evolved, the co-factor specificity was relaxed. Kinetic studies show that the T. maritima DHDPR enzyme is inhibited by high concentrations of its substrate, DHDP, and that at high concentrations NADH also acts as an inhibitor of the enzyme, suggesting a novel method of regulation for the lysine biosynthetic pathway. Increased thermal stability of the T. maritima DHDPR enzyme may be associated with the lack of C-terminal and N-terminal loops that are present in the E. coli DHDPR enzyme.


Subject(s)
Bacterial Proteins/chemistry , Dihydrodipicolinate Reductase/chemistry , Thermotoga maritima/enzymology , Bacterial Proteins/metabolism , Binding Sites , Biological Evolution , Dihydrodipicolinate Reductase/metabolism , Enzyme Stability , Kinetics , Models, Molecular , NAD/metabolism , NADP/metabolism , Protein Binding , Temperature
17.
Plant J ; 52(3): 539-47, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17727612

ABSTRACT

Chloroplast NAD(P)H dehydrogenase (NDH) is a homolog of the bacterial NADH dehydrogenase NDH-1 and is involved in cyclic electron transport around photosystem I. In higher plants, 14 subunits of the NDH complex have been identified. The subunit that contains the electron donor-binding site or an electron donor to NDH has not been determined. Arabidopsis crr1 (chlororespiratory reduction 1) mutants were isolated by chlorophyll fluorescence imaging on the basis of their lack of NDH activity. CRR1 is homologous to dihydrodipicolinate reductase (DHPR), which functions in a lysine biosynthesis pathway. However, the dihydrodipicolinate-binding motif was not conserved in CRR1, and the crr1 defect was specific to accumulation of the NDH complex, implying that CRR1 is not involved in lysine biosynthesis in Arabidopsis. Similarly to other nuclear-encoded genes for NDH subunits, CRR1 was expressed only in photosynthetic tissue. CRR1 contained a NAD(P)H-binding motif and was a candidate electron donor-binding subunit of the NDH complex. However, CRR1 was detected in the stroma but not in the thylakoid membranes, where the NDH complex is localized. Furthermore, CRR1 was stable in crr2-2 lacking the NDH complex. These results suggest that CRR1 is involved in biogenesis or stabilization of the NDH complex, possibly via the reduction of an unknown substrate.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Chloroplasts/enzymology , Dihydrodipicolinate Reductase/metabolism , NADPH Dehydrogenase/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Chloroplasts/metabolism , Dihydrodipicolinate Reductase/chemistry , Dihydrodipicolinate Reductase/genetics , Electron Transport , Gene Expression Regulation, Plant , Phylogeny , Plant Leaves/enzymology , Plant Leaves/genetics , Plant Leaves/metabolism , Sequence Alignment
18.
Microbiology (Reading) ; 152(Pt 1): 105-112, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16385120

ABSTRACT

The use of a lysine-overproducing strain of Lactobacillus plantarum in food or feed fermentations may lead to the production of lysine-rich products. The availability of functional genes and information on the regulation of lysine biosynthesis are required to develop a lysine-overproducing strain. The genome sequence of L. plantarum revealed putative lysine biosynthetic genes, some of which may produce isozymes. This study examined the functionality of the genes and the regulation of the first four enzymes of lysine biosynthesis, together with homoserine dehydrogenase, in L. plantarum. The genes were expressed in Escherichia coli, and the regulation of the enzymes was studied in cell extracts of both recombinant E. coli and L. plantarum. Among seven lysine biosynthetic genes studied (aspartokinase genes, thrA1 and thrA2; aspartate semialdehyde dehydrogenase genes, asd1 and asd2; dihydrodipicolinate synthase genes, dapA1 and dapA2; and the dihydrodipicolinate reductase gene, dapB) plus two homoserine dehydrogenase genes (hom1 and hom2), the products of six genes, i.e. thrA2, asd2, dapA1, dapB, hom1 and hom2, showed obvious enzyme activities in vitro. The product of one of the homoserine dehydrogenase genes, hom1, exhibited both homoserine dehydrogenase and aspartokinase activities. However, the aspartokinase activity was mainly due to ThrA2 and was inhibited by L-lysine and repressed by L-threonine, and the homoserine dehydrogenase activity was mainly due to Hom2 and was inhibited by L-threonine. The aspartate semialdehyde dehydrogenase, dihydrodipicolinate synthase and dihydrodipicolinate reductase were not regulated by the end-products of the pathway.


Subject(s)
Aspartate Kinase/metabolism , Aspartate-Semialdehyde Dehydrogenase/metabolism , Dihydrodipicolinate Reductase/metabolism , Hydro-Lyases/metabolism , Lactobacillus plantarum/metabolism , Aspartate Kinase/genetics , Aspartate-Semialdehyde Dehydrogenase/genetics , Dihydrodipicolinate Reductase/genetics , Gene Expression , Genome, Bacterial , Homoserine Dehydrogenase/genetics , Hydro-Lyases/genetics , Lactobacillus plantarum/genetics , Lysine/biosynthesis , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Threonine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...