Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Chembiochem ; 23(20): e202200285, 2022 10 19.
Article in English | MEDLINE | ID: mdl-35943842

ABSTRACT

Phosphonates are produced across all domains of life and used widely in medicine and agriculture. Biosynthesis almost universally originates from the enzyme phosphoenolpyruvate mutase (Ppm), EC 5.4.2.9, which catalyzes O-P bond cleavage in phosphoenolpyruvate (PEP) and forms a high energy C-P bond in phosphonopyruvate (PnPy). Mechanistic scrutiny of this unusual intramolecular O-to-C phosphoryl transfer began with the discovery of Ppm in 1988 and concluded in 2008 with computational evidence supporting a concerted phosphoryl transfer via a dissociative metaphosphate-like transition state. This mechanism deviates from the standard 'in-line attack' paradigm for enzymatic phosphoryl transfer that typically involves a phosphoryl-enzyme intermediate, but definitive evidence is sparse. Here we review the experimental evidence leading to our current mechanistic understanding and highlight the roles of previously underappreciated conserved active site residues. We then identify remaining opportunities to evaluate overlooked residues and unexamined substrates/inhibitors.


Subject(s)
Organophosphonates , Phosphotransferases (Phosphomutases) , Phosphoenolpyruvate/chemistry , Phosphotransferases (Phosphomutases)/chemistry , Catalysis
2.
Biochem J ; 477(11): 2095-2114, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32459324

ABSTRACT

Activation of phosphoenolpyruvate carboxylase (PEPC) enzymes by glucose 6-phosphate (G6P) and other phospho-sugars is of major physiological relevance. Previous kinetic, site-directed mutagenesis and crystallographic results are consistent with allosteric activation, but the existence of a G6P-allosteric site was questioned and competitive activation-in which G6P would bind to the active site eliciting the same positive homotropic effect as the substrate phosphoenolpyruvate (PEP)-was proposed. Here, we report the crystal structure of the PEPC-C4 isozyme from Zea mays with G6P well bound into the previously proposed allosteric site, unambiguously confirming its existence. To test its functionality, Asp239-which participates in a web of interactions of the protein with G6P-was changed to alanine. The D239A variant was not activated by G6P but, on the contrary, inhibited. Inhibition was also observed in the wild-type enzyme at concentrations of G6P higher than those producing activation, and probably arises from G6P binding to the active site in competition with PEP. The lower activity and cooperativity for the substrate PEP, lower activation by glycine and diminished response to malate of the D239A variant suggest that the heterotropic allosteric activation effects of free-PEP are also abolished in this variant. Together, our findings are consistent with both the existence of the G6P-allosteric site and its essentiality for the activation of PEPC enzymes by phosphorylated compounds. Furthermore, our findings suggest a central role of the G6P-allosteric site in the overall kinetics of these enzymes even in the absence of G6P or other phospho-sugars, because of its involvement in activation by free-PEP.


Subject(s)
Glucose-6-Phosphate/chemistry , Phosphoenolpyruvate Carboxylase/chemistry , Phosphoenolpyruvate/chemistry , Plant Proteins/chemistry , Zea mays/enzymology , Allosteric Regulation , Catalytic Domain , Glucose-6-Phosphate/metabolism , Kinetics , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate Carboxylase/genetics , Phosphoenolpyruvate Carboxylase/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Zea mays/genetics
3.
Arch Biochem Biophys ; 695: 108633, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33075302

ABSTRACT

A linked-function theory for allostery allows for a differentiation between those protein-ligand interactions that contribute the most to ligand binding and those protein-ligand interactions that contribute to the allosteric mechanism. This potential distinction is the basis for analogue studies used to determine which chemical moieties on the allosteric effector contribute to allostery. Although less recognized, the same separation of functions is possible for substrate-enzyme interactions. When evaluating allosteric regulation in human liver pyruvate kinase, the use of a range of monovalent cations (K+, NH4+, Rb+, Cs+, cyclohexylammonium+ and Tris+) altered substrate (phosphoenolpyruvate; PEP) affinity, but maintained similar allosteric responses to the allosteric activator, fructose-1,6-bisphosphate (Fru-1,6-BP). Because crystal structures indicate that the active site monovalent cation interacts directly with the phosphate moiety of the bound PEP substrate, we questioned if the phosphate moiety might contribute to substrate binding, but not to the allosteric mechanism. Here, we demonstrate that the binding of oxalate, a non-phosphorylated substrate/product analogue, is allosterically enhanced by Fru-1,6-BP. That observation is consistent with the concept that the phosphate moiety of PEP is not required for the allosteric function, even though that moiety likely contributes to determining substrate affinity.


Subject(s)
Fructosediphosphates/chemistry , Liver/enzymology , Phosphoenolpyruvate/chemistry , Pyruvate Kinase/chemistry , Allosteric Regulation , Fructosediphosphates/metabolism , Humans , Phosphoenolpyruvate/metabolism , Pyruvate Kinase/metabolism
4.
Biochemistry ; 58(35): 3669-3682, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31386812

ABSTRACT

Cancer cells regulate key enzymes in the glycolytic pathway to control the glycolytic flux, which is necessary for their growth and proliferation. One of the enzymes is pyruvate kinase muscle isoform 2 (PKM2), which is allosterically regulated by various small molecules. Using detailed biochemical and kinetic studies, we demonstrate that cysteine inhibits wild-type (wt) PKM2 by shifting from an active tetramer to a mixture of a tetramer and a less active dimer/monomer equilibrium and that the inhibition is dependent on cysteine concentration. The cysteine-mediated PKM2 inhibition is reversed by fructose 1,6-bisphosphate, an allosteric activator of PKM2. Furthermore, kinetic studies using two dimeric PKM2 variants, S437Y PKM2 and G415R PKM2, show that the reversal is caused by the tetramerization of wtPKM2. The crystal structure of the wtPKM2-Cys complex was determined at 2.25 Å, which showed that cysteine is held to the amino acid binding site via its main chain groups, similar to that observed for phenylalanine, alanine, serine, and tryptophan. Notably, ligand binding studies using fluorescence and isothermal titration calorimetry show that the presence of phosphoenolpyruvate alters the binding affinities of amino acids for wtPKM2 and vice versa, thereby unravelling the existence of a functionally bidirectional coupling between the amino acid binding site and the active site of wtPKM2.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Cysteine/pharmacology , Enzyme Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Thyroid Hormones/chemistry , Amino Acid Substitution/genetics , Amino Acids/chemistry , Amino Acids/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Catalysis , Catalytic Domain/genetics , Crystallography, X-Ray , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphoenolpyruvate/chemistry , Phosphoenolpyruvate/metabolism , Protein Binding , Protein Conformation , Serine/genetics , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Tyrosine/genetics , Thyroid Hormone-Binding Proteins
5.
Biochemistry ; 56(4): 592-601, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28045507

ABSTRACT

3-Deoxy-d-arabino-heptulosonate-7-phosphate (DAHP) synthase catalyzes an aldol-like reaction of phosphoenolpyruvate (PEP) with erythrose 4-phosphate (E4P) to form DAHP in the first step of the shikimate biosynthetic pathway. DAHP oxime, in which an oxime replaces the ketone, is a potent inhibitor, with Ki = 1.5 µM. Linear free energy relationship (LFER) analysis of DAHP oxime inhibition using DAHP synthase mutants revealed an excellent correlation between transition state stabilization and inhibition. The equations of LFER analysis were rederived to formalize the possibility of proportional, rather than equal, changes in the free energies of transition state stabilization and inhibitor binding, in accord with the fact that the majority of LFER analyses in the literature demonstrate nonunity slopes. A slope of unity, m = 1, indicates that catalysis and inhibitor binding are equally sensitive to perturbations such as mutations or modified inhibitor/substrate structures. Slopes <1 or >1 indicate that inhibitor binding is less sensitive or more sensitive, respectively, to perturbations than is catalysis. LFER analysis using the tetramolecular specificity constant, that is, plotting log(KM,MnKM,PEPKM,E4P/kcat) versus log(Ki), revealed a slope, m, of 0.34, with r2 = 0.93. This provides evidence that DAHP oxime is mimicking the first irreversible transition state of the DAHP synthase reaction, presumably phosphate departure from the tetrahedral intermediate. This is evidence that the oxime group can act as a functional, as well as structural, mimic of phosphate groups.


Subject(s)
3-Deoxy-7-Phosphoheptulonate Synthase/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Oximes/chemistry , Recombinant Fusion Proteins/chemistry , Sugar Phosphates/chemistry , 3-Deoxy-7-Phosphoheptulonate Synthase/chemistry , 3-Deoxy-7-Phosphoheptulonate Synthase/genetics , 3-Deoxy-7-Phosphoheptulonate Synthase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biocatalysis , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Kinetics , Models, Molecular , Molecular Mimicry , Mutation , Phosphoenolpyruvate/chemistry , Phosphoenolpyruvate/metabolism , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Shikimic Acid/chemistry , Shikimic Acid/metabolism , Sugar Phosphates/metabolism , Thermodynamics
6.
Arch Biochem Biophys ; 607: 1-6, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27477958

ABSTRACT

The allosteric coupling free energy between ligands fructose-6-phosphate (Fru-6-P) and phospho(enol)pyruvate (PEP) for phosphofructokinase-1 (PFK) from the moderate thermophile, Bacillus stearothermophilus (BsPFK), results from compensating enthalpy and entropy components. In BsPFK the positive coupling free energy that defines inhibition is opposite in sign from the negative enthalpy term and is therefore determined by the larger absolute value of the negative entropy term. Variants of BsPFK were made to determine the effect of adding small cavities to the structure on the allosteric function of the enzyme. The BsPFK Ile → Val (cavity containing) mutants have varied values for the coupling free energy between PEP and Fru-6-P, indicating that the modifications altered the effectiveness of PEP as an inhibitor. Notably, the mutation I153V had a substantial positive impact on the magnitude of inhibition by PEP. Van't Hoff analysis determined that this is the result of decreased entropy-enthalpy compensation with a larger change in the enthalpy term compared to the entropy term.


Subject(s)
Bacterial Proteins/chemistry , Geobacillus stearothermophilus/enzymology , Phosphofructokinases/chemistry , Allosteric Site , Bacterial Proteins/genetics , Catalysis , Crystallography, X-Ray , Fructosephosphates/chemistry , Geobacillus stearothermophilus/genetics , Hydrogen-Ion Concentration , Kinetics , Molecular Conformation , Mutagenesis, Site-Directed , Mutation , Phosphoenolpyruvate/chemistry , Phosphofructokinases/genetics , Temperature
7.
Arch Biochem Biophys ; 606: 26-33, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27431058

ABSTRACT

Although oxidative stress is known to impede the tricarboxylic acid (TCA) cycle and oxidative phosphorylation, the nutritionally-versatile microbe, Pseudomonas fluorescens has been shown to proliferate in the presence of hydrogen peroxide (H2O2) and nitrosative stress. In this study we demonstrate the phospho-transfer system that enables this organism to generate ATP was similar irrespective of the carbon source utilized. Despite the diminished activities of enzymes involved in the TCA cycle and in the electron transport chain (ETC), the ATP levels did not appear to be significantly affected in the stressed cells. Phospho-transfer networks mediated by acetate kinase (ACK), adenylate kinase (AK), and nucleoside diphosphate kinase (NDPK) are involved in maintaining ATP homeostasis in the oxidatively-challenged cells. This phospho-relay machinery orchestrated by substrate-level phosphorylation is aided by the up-regulation in the activities of such enzymes like phosphoenolpyruvate carboxylase (PEPC), pyruvate orthophosphate dikinase (PPDK), and phosphoenolpyruvate synthase (PEPS). The enhanced production of phosphoenolpyruvate (PEP) and pyruvate further fuel the synthesis of ATP. Taken together, this metabolic reconfiguration enables the organism to fulfill its ATP need in an O2-independent manner by utilizing an intricate phospho-wire module aimed at maximizing the energy potential of PEP with the participation of AMP.


Subject(s)
Adenosine Triphosphate/chemistry , Pseudomonas fluorescens/metabolism , Adenosine Monophosphate/chemistry , Citric Acid Cycle , Densitometry , Electron Transport , Homeostasis , Hydrogen Peroxide/chemistry , Lipids/chemistry , Oxidation-Reduction , Oxidative Phosphorylation , Oxidative Stress , Oxygen/chemistry , Phosphoenolpyruvate/chemistry , Phosphorylation , Phosphotransferases (Paired Acceptors)/metabolism , Pyruvate, Orthophosphate Dikinase/metabolism , Reactive Oxygen Species/metabolism
8.
Glycobiology ; 25(1): 115-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25214154

ABSTRACT

N-Acetylneuraminate lyase synthase (NeuB; E.C. 2.5.1.56) is a key enzyme in pathogenic microorganisms for producing N-acetylneuraminic acid through the irreversible condensation of N-acetylmannosamine (ManNAc) and phosphoenolpyruvate (PEP). However, nothing is known about this enzyme in non-pathogenic bacteria. This paper describes, for the first time, one of the two putative N-acetylneuraminate synthases from the halophilic non-pathogenic gamma-proteobacterium Idiomarina loihiensis NeuB1 (IlNeuB1). The obtained 95-kDa dimeric enzyme showed maximal activity at pH 7.0 and 40°C and was more stable at pH 8.0 (8 h half-life) than the previously described NeuB. Its catalytic efficiency towards ManNAc and PEP was 10- and 40-fold higher, respectively, than that determined for Campylobacter jejuni NeuB, but only half that found for Neisseria meningitidis NeuB towards PEP. The phylogenetic and structural analyses of NeuB enzymes revealed the new domain architecture 4 has no cystathionine-ß-synthase domain (cystathionine-ß-synthetase domain), unlike domain architecture 3. In addition, 10 conserved blocks (I-X) were found, and surprisingly, this study showed that the arginine essential for catalysis that is present in antifreeze-like domain (block X) was not fully conserved in NeuB, but is replaced by a serine in a long sequence (>700 residues) NeuB, such as that existing in domain architectures 3 and 4.


Subject(s)
Alteromonadaceae/chemistry , Bacterial Proteins/chemistry , Hexosamines/chemistry , Oxo-Acid-Lyases/chemistry , Phosphoenolpyruvate/chemistry , Alteromonadaceae/classification , Alteromonadaceae/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Campylobacter jejuni/chemistry , Campylobacter jejuni/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Half-Life , Hexosamines/metabolism , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Neisseria meningitidis/chemistry , Neisseria meningitidis/enzymology , Oxo-Acid-Lyases/genetics , Oxo-Acid-Lyases/metabolism , Phosphoenolpyruvate/metabolism , Phylogeny , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Species Specificity , Substrate Specificity
9.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 12): 2457-70, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26627653

ABSTRACT

Staphylococcus aureus is a Gram-positive bacterium with strong pathogenicity that causes a wide range of infections and diseases. Enolase is an evolutionarily conserved enzyme that plays a key role in energy production through glycolysis. Additionally, enolase is located on the surface of S. aureus and is involved in processes leading to infection. Here, crystal structures of Sa_enolase with and without bound phosphoenolpyruvate (PEP) are presented at 1.6 and 2.45 Šresolution, respectively. The structure reveals an octameric arrangement; however, both dimeric and octameric conformations were observed in solution. Furthermore, enzyme-activity assays show that only the octameric variant is catalytically active. Biochemical and structural studies indicate that the octameric form of Sa_enolase is enzymatically active in vitro and likely also in vivo, while the dimeric form is catalytically inactive and may be involved in other biological processes.


Subject(s)
Bacterial Proteins/chemistry , Phosphoenolpyruvate/chemistry , Phosphopyruvate Hydratase/chemistry , Staphylococcus aureus/chemistry , Amino Acid Motifs , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Models, Molecular , Molecular Sequence Data , Phosphoenolpyruvate/metabolism , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staphylococcus aureus/enzymology
10.
Protein Expr Purif ; 110: 7-13, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25573389

ABSTRACT

The cDNA encoding for a Solanum tuberosum cytosolic pyruvate kinase 1 (PKc1) highly expressed in tuber tissue was cloned in the bacterial expression vector pProEX HTc. The construct carried a hexahistidine tag in N-terminal position to facilitate purification of the recombinant protein. Production of high levels of soluble recombinant PKc1 in Escherichia coli was only possible when using a co-expression strategy with the chaperones GroES-GroEL. Purification of the protein by Ni(2 +) chelation chromatography yielded a single protein with an apparent molecular mass of 58kDa and a specific activity of 34unitsmg(-1) protein. The recombinant enzyme had an optimum pH between 6 and 7. It was relatively heat stable as it retained 80% of its activity after 2min at 75°C. Hyperbolic saturation kinetics were observed with ADP and UDP whereas sigmoidal saturation was observed during analysis of phosphoenolpyruvate binding. Among possible effectors tested, aspartate and glutamate had no effect on enzyme activity, whereas α-ketoglutarate and citrate were the most potent inhibitors. When tested on phosphoenolpyruvate saturation kinetics, these latter compounds increased S0.5. These findings suggest that S. tuberosum PKc1 is subject to a strong control by respiratory metabolism exerted via citrate and other tricarboxylic acid cycle intermediates.


Subject(s)
Cytosol/chemistry , Phosphoenolpyruvate/chemistry , Plant Proteins/isolation & purification , Pyruvate Kinase/isolation & purification , Solanum tuberosum/chemistry , Adenosine Diphosphate/chemistry , Citric Acid/chemistry , Cloning, Molecular , Cytosol/enzymology , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hydrogen-Ion Concentration , Ketoglutaric Acids/chemistry , Kinetics , Molecular Weight , Plant Proteins/antagonists & inhibitors , Plant Proteins/biosynthesis , Plant Proteins/genetics , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/biosynthesis , Pyruvate Kinase/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Solanum tuberosum/enzymology , Uridine Diphosphate/chemistry
11.
Int J Mol Sci ; 16(7): 14490-510, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26121302

ABSTRACT

Streptococcus iniae is a major fish pathogen that can also cause human bacteremia, cellulitis and meningitis. Screening for and identification of protective antigens plays an important role in developing therapies against S. iniae infections. In this study, we indicated that the α-enolase of S. iniae was not only distributed in the cytoplasm and associated to cell walls, but was also secreted to the bacterial cell surface. The functional identity of the purified recombinant α-enolase protein was verified by its ability to catalyze the conversion of 2-phosphoglycerate (2-PGE) to phosphoenolpyruvate (PEP), and both the recombinant and native proteins interacted with human plasminogen. The rabbit anti-rENO serum blockade assay shows that α-enolase participates in S. iniae adhesion to and invasion of BHK-21 cells. In addition, the recombinant α-enolase can confer effective protection against S. iniae infection in mice, which suggests that α-enolase has potential as a vaccine candidate in mammals. We conclude that S. iniae α-enolase is a moonlighting protein that also associates with the bacterial outer surface and functions as a protective antigen in mice.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/metabolism , Phosphopyruvate Hydratase/metabolism , Streptococcus/enzymology , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Cell Adhesion , Cell Line , Cell Movement , Cloning, Molecular , Cricetinae , Cricetulus , Glyceric Acids/chemistry , Mice , Molecular Sequence Data , Phosphoenolpyruvate/chemistry , Phosphopyruvate Hydratase/chemistry , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/immunology , Streptococcus/genetics , Streptococcus/immunology
12.
Proteins ; 82(9): 2054-66, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24633984

ABSTRACT

N-Acetylneuraminic acid (NANA) is the most common naturally occurring sialic acid and plays a key role in the pathogenesis of a select number of neuroinvasive bacteria such as Neisseria meningitidis. NANA is synthesized in prokaryotes via a condensation reaction between phosphoenolpyruvate and N-acetylmannosamine. This reaction is catalyzed by a domain swapped, homodimeric enzyme, N-acetylneuraminic acid synthase (NANAS). NANAS comprises two distinct domains; an N-terminal catalytic (ß/α)8 barrel linked to a C-terminal antifreeze protein-like (AFPL) domain. We have investigated the role of the AFPL domain by characterizing a truncated variant of NmeNANAS, which was discovered to be soluble yet inactive. Analytical ultracentrifugation and analytical size exclusion were used to probe the quaternary state of the NmeNANAS truncation, and revealed that loss of the AFPL domain destabilizes the dimeric form of the enzyme. The results from this study thereby demonstrate that the AFPL domain plays a critical role for both the catalytic function and quaternary structure stability of NANAS. Small angle X-ray scattering, molecular dynamics simulations, and amino acid substitutions expose a complex hydrogen-bonding relay, which links the roles of the catalytic and AFPL domains across subunit boundaries.


Subject(s)
Catalytic Domain , Neisseria meningitidis/pathogenicity , Oxo-Acid-Lyases/metabolism , Binding Sites , Chromatography, Gel , Crystallography, X-Ray , Hexosamines/chemistry , Hydrogen Bonding , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , N-Acetylneuraminic Acid/biosynthesis , Phosphoenolpyruvate/chemistry , Protein Structure, Quaternary
13.
Microb Cell Fact ; 13(1): 28, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24559297

ABSTRACT

BACKGROUND: Efficient production of SA in Escherichia coli has been achieved by modifying key genes of the central carbon metabolism and SA pathway, resulting in overproducing strains grown in batch- or fed-batch-fermentor cultures using a complex broth including glucose and YE. In this study, we performed a GTA to identify those genes significantly upregulated in an engineered E. coli strain, PB12.SA22, in mid EXP (5 h), early STA (STA1, 9 h), and late STA (STA2, 44 h) phases, grown in complex fermentation broth in batch-fermentor cultures. RESULTS: Growth of E. coli PB12.SA22 in complex fermentation broth for SA production resulted in an EXP growth during the first 9 h of cultivation depending of supernatant available aromatic amino acids provided by YE because, when tryptophan was totally consumed, cells entered into a second, low-growth phase (even in the presence of glucose) until 26 h of cultivation. At this point, glucose was completely consumed but SA production continued until the end of the fermentation (50 h) achieving the highest accumulation (7.63 g/L of SA). GTA between EXP/STA1, EXP/STA2 and STA1/STA2 comparisons showed no significant differences in the regulation of genes encoding enzymes of central carbon metabolism as in SA pathway, but those genes encoding enzymes involved in sugar, amino acid, nucleotide/nucleoside, iron and sulfur transport; amino acid catabolism and biosynthesis; nucleotide/nucleoside salvage; acid stress response and modification of IM and OM were upregulated between comparisons. CONCLUSIONS: GTA during SA production in batch-fermentor cultures of strain PB12.SA22 grown in complex fermentation broth during the EXP, STA1 and STA2 phases was studied. Significantly, upregulated genes during the EXP and STA1 phases were associated with transport, amino acid catabolism, biosynthesis, and nucleotide/nucleoside salvage. In STA2, upregulation of genes encoding transporters and enzymes involved in the synthesis and catabolism of Arg suggests that this amino acid could have a key role in the fuelling of carbon toward SA synthesis, whereas upregulation of genes involved in pH stress response, such as membrane modifications, suggests a possible response to environmental conditions imposed on the cell at the end of the fermentation.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Phosphoenolpyruvate Sugar Phosphotransferase System/metabolism , Phosphoenolpyruvate/chemistry , Shikimic Acid/metabolism , Amino Acids/metabolism , Batch Cell Culture Techniques , Culture Media/chemistry , Escherichia coli/genetics , Gene Expression Profiling , Gene Regulatory Networks , Genetic Engineering , Up-Regulation
14.
Anal Bioanal Chem ; 406(14): 3365-70, 2014 May.
Article in English | MEDLINE | ID: mdl-24748446

ABSTRACT

The biocatalytic cascade based on enzyme-catalyzed reactions activated by several biomolecular input signals and producing output signal after each reaction step was developed as an example of a logically reversible information processing system. The model system was designed to mimic the operation of concatenated AND logic gates with optically readable output signals generated at each step of the logic operation. Implications include concurrent bioanalyses and data interpretation for medical diagnostics.


Subject(s)
Biological Assay/instrumentation , Biosensing Techniques , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Biological Assay/methods , Biomarkers/chemistry , Catalysis , Chemistry Techniques, Analytical , Diagnostic Tests, Routine , Glyceric Acids/chemistry , Humans , L-Lactate Dehydrogenase/chemistry , Mixed Function Oxygenases/chemistry , NAD/chemistry , Optics and Photonics , Oxygen/chemistry , Phosphoenolpyruvate/chemistry , Phosphopyruvate Hydratase/chemistry
15.
Proc Natl Acad Sci U S A ; 108(36): 14763-8, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21873244

ABSTRACT

Bioavailable calcium is maintained by some crustaceans, in particular freshwater crayfish, by stabilizing amorphous calcium carbonate (ACC) within reservoir organs--gastroliths, readily providing the Ca(2+) needed to build a new exoskeleton. Despite the key scientific and biomedical importance of the in situ molecular-level picture of biogenic ACC and its stabilization in a bioavailable form, its description has eluded efforts to date. Herein, using multinuclear NMR, we accomplish in situ molecular-level characterization of ACC within intact gastroliths of the crayfish Cherax quadricarinatus. In addition to the known CaCO(3), chitin scaffold and inorganic phosphate (Pi), we identify within the gastrolith two primary metabolites, citrate and phosphoenolpyruvate (PEP) and quantify their abundance by applying solution NMR techniques to the gastrolith "soluble matrix." The long-standing question on the physico-chemical state of ACC stabilizing, P-bearing moieties within the gastrolith is answered directly by the application of solid state rotational-echo double-resonance (REDOR) and transferred-echo double-resonance (TEDOR) NMR to the intact gastroliths: Pi and PEP are found molecularly dispersed throughout the ACC as a solid solution. Citrate carboxylates are found < 5 Å from a phosphate (intermolecular CP distance), an interaction that must be mediated by Ca(2+). The high abundance and extensive interactions of these molecules with the ACC matrix identify them as the central constituents stabilizing the bioavailable form of calcium. This study further emphasizes that it is imperative to characterize the intact biogenic CaCO(3). Solid state NMR spectroscopy is shown to be a robust and accessible means of determining composition, internal structure, and molecular functionality in situ.


Subject(s)
Astacoidea/chemistry , Calcium Carbonate/chemistry , Chitin/chemistry , Citrates/chemistry , Phosphoenolpyruvate/chemistry , Animals , Astacoidea/metabolism , Calcium Carbonate/metabolism , Chitin/metabolism , Citrates/metabolism , Magnetic Resonance Spectroscopy , Phosphoenolpyruvate/metabolism
16.
J Mol Biol ; 436(9): 168553, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38548260

ABSTRACT

The catalytic cycle of Enzyme I (EI), a phosphotransferase enzyme responsible for converting phosphoenolpyruvate (PEP) into pyruvate, is characterized by a series of local and global conformational rearrangements. This multistep process includes a monomer-to-dimer transition, followed by an open-to-closed rearrangement of the dimeric complex upon PEP binding. In the present study, we investigate the thermodynamics of EI dimerization using a range of high-pressure solution NMR techniques complemented by SAXS experiments. 1H-15N TROSY and 1H-13C methyl TROSY NMR spectra combined with 15N relaxation measurements revealed that a native-like engineered variant of full-length EI fully dissociates into stable monomeric state above 1.5 kbar. Conformational ensembles of EI monomeric state were generated via a recently developed protocol combining coarse-grained molecular simulations with experimental backbone residual dipolar coupling measurements. Analysis of the structural ensembles provided detailed insights into the molecular mechanisms driving formation of the catalytically competent dimeric state, and reveals that each step of EI catalytical cycle is associated with a significant reduction in either inter- or intra-domain conformational entropy. Altogether, this study completes a large body work conducted by our group on EI and establishes a comprehensive structural and dynamical description of the catalytic cycle of this prototypical multidomain, oligomeric enzyme.


Subject(s)
Phosphoenolpyruvate Sugar Phosphotransferase System , Phosphotransferases (Nitrogenous Group Acceptor) , Protein Multimerization , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate/chemistry , Phosphoenolpyruvate Sugar Phosphotransferase System/chemistry , Phosphotransferases (Nitrogenous Group Acceptor)/chemistry , Protein Conformation , Scattering, Small Angle , Thermodynamics , X-Ray Diffraction
17.
Talanta ; 275: 126134, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38692044

ABSTRACT

Phosphoenolpyruvate (PEP) is an essential intermediate metabolite that is involved in various vital biochemical reactions. However, achieving the direct and accurate quantification of PEP in plasma or serum poses a significant challenge owing to its strong polarity and metal affinity. In this study, a sensitive method for the direct determination of PEP in plasma and serum based on ethylenediaminetetraacetic acid (EDTA)-facilitated hydrophilic interaction liquid chromatography-tandem mass spectrometry was developed. Superior chromatographic retention and peak shapes were achieved using a zwitterionic stationary-phase HILIC column with a metal-inert inner surface. Efficient dechelation of PEP-metal complexes in serum/plasma samples was achieved through the introduction of EDTA, resulting in a significant enhancement of the PEP signal. A PEP isotopically labelled standard was employed as a surrogate analyte for the determination of endogenous PEP, and validation assessments proved the sensitivity, selectivity, and reproducibility of this method. The method was applied to the comparative quantification of PEP in plasma and serum samples from mice and rats, as well as in HepG2 cells, HEK293T cells, and erythrocytes; the results confirmed its applicability in PEP-related biomedical research. The developed method can quantify PEP in diverse biological matrices, providing a feasible opportunity to investigate the role of PEP in relevant biomedical research.


Subject(s)
Edetic Acid , Hydrophobic and Hydrophilic Interactions , Phosphoenolpyruvate , Tandem Mass Spectrometry , Tandem Mass Spectrometry/methods , Animals , Humans , Edetic Acid/chemistry , Mice , Chromatography, Liquid/methods , Rats , Phosphoenolpyruvate/chemistry , Phosphoenolpyruvate/blood , Phosphoenolpyruvate/metabolism , HEK293 Cells , Hep G2 Cells , Rats, Sprague-Dawley , Male
18.
Protein Sci ; 33(7): e5075, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38895978

ABSTRACT

Rheostat positions, which can be substituted with various amino acids to tune protein function across a range of outcomes, are a developing area for advancing personalized medicine and bioengineering. Current methods cannot accurately predict which proteins contain rheostat positions or their substitution outcomes. To compare the prevalence of rheostat positions in homologs, we previously investigated their occurrence in two pyruvate kinase (PYK) isozymes. Human liver PYK contained numerous rheostat positions that tuned the apparent affinity for the substrate phosphoenolpyruvate (Kapp-PEP) across a wide range. In contrast, no functional rheostat positions were identified in Zymomonas mobilis PYK (ZmPYK). Further, the set of ZmPYK substitutions included an unusually large number that lacked measurable activity. We hypothesized that the inactive substitution variants had reduced protein stability, precluding detection of Kapp-PEP tuning. Using modified buffers, robust enzymatic activity was obtained for 19 previously-inactive ZmPYK substitution variants at three positions. Surprisingly, both previously-inactive and previously-active substitution variants all had Kapp-PEP values close to wild-type. Thus, none of the three positions were functional rheostat positions, and, unlike human liver PYK, ZmPYK's Kapp-PEP remained poorly tunable by single substitutions. To directly assess effects on stability, we performed thermal denaturation experiments for all ZmPYK substitution variants. Many diminished stability, two enhanced stability, and the three positions showed different thermal sensitivity to substitution, with one position acting as a "stability rheostat." The differences between the two PYK homologs raises interesting questions about the underlying mechanism(s) that permit functional tuning by single substitutions in some proteins but not in others.


Subject(s)
Pyruvate Kinase , Zymomonas , Humans , Zymomonas/enzymology , Zymomonas/genetics , Zymomonas/chemistry , Zymomonas/metabolism , Pyruvate Kinase/chemistry , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Amino Acid Substitution , Protein Stability , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Enzyme Stability , Liver/enzymology , Liver/metabolism , Liver/chemistry , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate/chemistry
19.
Biochemistry ; 52(1): 1-3, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23256782

ABSTRACT

A series of substrate analogues has been used to determine which chemical moieties of the substrate phosphoenolpyruvate (PEP) contribute to the allosteric inhibition of rabbit muscle pyruvate kinase by phenylalanine. Replacing the carboxyl group of the substrate with a methyl alcohol or removing the phosphate altogether greatly reduces substrate affinity. However, removal of the carboxyl group is the only modification tested that removes the ability to allosterically reduce the level of Phe binding. From this, it can be concluded that the carboxyl group of PEP is responsible for energetic coupling with Phe binding in the allosteric sites.


Subject(s)
Muscles/enzymology , Phenylalanine/metabolism , Phosphoenolpyruvate/chemistry , Phosphoenolpyruvate/metabolism , Pyruvate Kinase/metabolism , Allosteric Regulation , Allosteric Site , Animals , Muscles/chemistry , Phosphoenolpyruvate/analogs & derivatives , Protein Binding , Pyruvate Kinase/chemistry , Rabbits , Substrate Specificity
20.
Biochemistry ; 52(32): 5421-9, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23859543

ABSTRACT

Bacillus stearothermophilus phosphofructokinase (BsPFK) is a homotetramer that is allosterically inhibited by phosphoenolpyruvate (PEP), which binds along one dimer-dimer interface. The substrate, fructose 6-phosphate (Fru-6-P), binds along the other dimer-dimer interface. Evans et al. observed that the structure with inhibitor (phosphoglycolate) bound, compared to the structure of wild-type BsPFK with substrate and activator bound, exhibits a 7° rotation about the substrate-binding interface, termed the quaternary shift [Schirmer, T., and Evans, P. R. (1990) Nature 343, 140-145]. We report that the variant D12A BsPFK exhibits a 100-fold increase in its binding affinity for PEP, a 50-fold decrease in its binding affinity for Fru-6-P, but an inhibitory coupling comparable to that of the wild type. Crystal structures of the apo and PEP-bound forms of D12A BsPFK have been determined (Protein Data Bank entries 4I36 and 4I7E , respectively), and both indicate a shifted structure similar to the inhibitor-bound structure of the wild type. D12 does not directly bind to either substrate or inhibitor and is located along the substrate-binding interface. A conserved hydrogen bond between D12 and T156 forms across the substrate-binding subunit-subunit interface in the substrate-bound form of BsPFK. The variant T156A BsPFK, when compared to the wild type, shows a 30-fold increase in PEP binding affinity, a 17-fold decrease in Fru-6-P binding affinity, and an estimated coupling that is also approximately equal to that of the wild type. In addition, the T156A BsPFK crystal structure bound to PEP is reported (Protein Data Bank entry 4I4I ), and it exhibits a shifted structure similar to that of D12A BsPFK and the inhibitor-bound structure of the wild type. The results suggest that the main role of the quaternary shift may be to influence ligand binding and not to cause the heterotropic allosteric inhibition per se.


Subject(s)
Bacterial Proteins/chemistry , Geobacillus stearothermophilus/enzymology , Phosphofructokinases/chemistry , Allosteric Regulation , Allosteric Site , Bacterial Proteins/metabolism , Binding Sites , Fructosephosphates/chemistry , Fructosephosphates/metabolism , Geobacillus stearothermophilus/metabolism , Hydrogen Bonding , Kinetics , Ligands , Phosphoenolpyruvate/chemistry , Phosphoenolpyruvate/metabolism , Phosphofructokinases/metabolism , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL