Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
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
2.
Biochim Biophys Acta Proteins Proteom ; 1868(2): 140331, 2020 02.
Article in English | MEDLINE | ID: mdl-31760039

ABSTRACT

In general, eukaryotic glucose-6-phosphate dehydrogenases (G6PDHs) are structurally stabilized by NADP+. Here we show by spectrofluorometric analysis, thermal and urea denaturation, and trypsin proteolysis, that a different mechanism stabilizes the enzyme from Pseudomonas aeruginosa (PaG6PDH) (EC 1.1.1.363). The spectrofluorometric analysis of the emission of 8-anilino-1-naphthalenesulfonic acid (ANS) indicates that this stabilization is the result of a structural change in the enzyme caused by G6P. The similarity between the Kd values determined for the PaG6PDH-G6P complex (78.0 ±â€¯7.9 µM) and the K0.5 values determined for G6P (57.9 ±â€¯2.5 and 104.5 ±â€¯9.3 µM in the NADP+- and NAD+-dependent reactions, respectively) suggests that the structural changes are the result of G6P binding to the active site of PaG6PDH. Modeling of PaG6PDH indicated the residues that potentially bind the ligand. These results and a phylogenetic analysis of the amino acid sequences of forty-four G6PDHs, suggest that the stabilization observed for PaG6PDH could be a characteristic that distinguishes this and other G6PDHs that use NAD+ and NADP+ from those that use NADP+ only or preferentially, such as those found in eukaryotes. This characteristic could be related to the metabolic roles these enzymes play in the organisms to which they belong.


Subject(s)
Glucosephosphate Dehydrogenase/metabolism , Pseudomonas aeruginosa/enzymology , Amino Acid Sequence , Anilino Naphthalenesulfonates/chemistry , Binding Sites , Catalytic Domain , Glucose-6-Phosphate/chemistry , Glucose-6-Phosphate/metabolism , Glucosephosphate Dehydrogenase/classification , Glucosephosphate Dehydrogenase/genetics , Kinetics , Molecular Dynamics Simulation , NAD/metabolism , NADP/chemistry , NADP/metabolism , Phylogeny , Protein Binding , Protein Denaturation , Protein Stability , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
3.
Arch Biochem Biophys ; 580: 50-6, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26116788

ABSTRACT

Optimization of cellulose enzymatic hydrolysis is crucial for cost effective bioethanol production from lignocellulosic biomass. Enzymes involved in cellulose hydrolysis are often inhibited by their end-products, cellobiose and glucose. Efforts have been made to produce more efficient enzyme variants that are highly tolerant to product accumulation; however, further improvements are still necessary. Based on an alternative approach we initially investigated whether recently formed glucose could be phosphorylated into glucose-6-phosphate to circumvent glucose accumulation and avoid inhibition of beta-glucosidase from Bacillus polymyxa (BGLA). The kinetic properties and structural analysis of BGLA in the presence of glucose-6-phosphate (G6P) were investigated. Kinetic studies demonstrated that enzyme was not inhibited by G6P. In contrast, the presence of G6P activated the enzyme, prevented beta glucosidase feedback inhibition by glucose accumulation and improved protein stability. G6P binding was investigated by fluorescence quenching experiments and the respective association constant indicated high affinity binding of G6P to BGLA. Data reported here are of great impact for future design strategies for second-generation bioethanol production.


Subject(s)
Bacillus/chemistry , Bacterial Proteins/chemistry , Glucose-6-Phosphate/chemistry , beta-Glucosidase/chemistry , Bacillus/enzymology , Bacterial Proteins/genetics , Enzyme Activation , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Glucose/chemistry , Kinetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thermodynamics , beta-Glucosidase/genetics
4.
FEBS J ; 279(13): 2296-309, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22519976

ABSTRACT

In Escherichia coli, the pentose phosphate pathway is one of the main sources of NADPH. The first enzyme of the pathway, glucose-6-phosphate dehydrogenase (G6PDH), is generally considered an exclusive NADPH producer, but a rigorous assessment of cofactor preference has yet to be reported. In this work, the specificity constants for NADP and NAD for G6PDH were determined using a pure enzyme preparation. Absence of the phosphate group on the cofactor leads to a 410-fold reduction in the performance of the enzyme. Furthermore, the contribution of the phosphate group to binding of the transition state to the active site was calculated to be 3.6 kcal·mol(-1). In order to estimate the main kinetic parameters for NAD(P) and NAD(P)H, we used the classical initial-rates approach, together with an analysis of reaction time courses. To achieve this, we developed a new analytical solution to the integrated Michaelis-Menten equation by including the effect of competitive product inhibition using the ω-function. With reference to relevant kinetic parameters and intracellular metabolite concentrations reported by others, we modeled the sensitivity of reduced cofactor production by G6PDH as a function of the redox ratios of NAD/NADH (rR(NAD)) and NADP/NADPH (rR(NADP)). Our analysis shows that NADPH production sharply increases within the range of thermodynamically feasible values of rR(NADP), but NADH production remains low within the range feasible for rR(NAD). Nevertheless, we show that certain combinations of rR(NADP) and rR(NAD) sustain greater levels of NADH production over NADPH.


Subject(s)
Escherichia coli/enzymology , Glucose-6-Phosphate/chemistry , Glucosephosphate Dehydrogenase/metabolism , Models, Biological , NADP/metabolism , NAD/metabolism , Binding, Competitive , Glucose-6-Phosphate/metabolism , Kinetics , Oxidation-Reduction
5.
FEBS J ; 273(9): 1975-88, 2006 May.
Article in English | MEDLINE | ID: mdl-16640561

ABSTRACT

Control analysis of the glycolytic flux was carried out in two fast-growth tumor cell types of human and rodent origin (HeLa and AS-30D, respectively). Determination of the maximal velocity (V(max)) of the 10 glycolytic enzymes from hexokinase to lactate dehydrogenase revealed that hexokinase (153-306 times) and phosphofructokinase-1 (PFK-1) (22-56 times) had higher over-expression in rat AS-30D hepatoma cells than in normal freshly isolated rat hepatocytes. Moreover, the steady-state concentrations of the glycolytic metabolites, particularly those of the products of hexokinase and PFK-1, were increased compared with hepatocytes. In HeLa cells, V(max) values and metabolite concentrations for the 10 glycolytic enzyme were also significantly increased, but to a much lesser extent (6-9 times for both hexokinase and PFK-1). Elasticity-based analysis of the glycolytic flux in AS-30D cells showed that the block of enzymes producing Fru(1,6)P2 (i.e. glucose transporter, hexokinase, hexosephosphate isomerase, PFK-1, and the Glc6P branches) exerted most of the flux control (70-75%), whereas the consuming block (from aldolase to lactate dehydrogenase) exhibited the remaining control. The Glc6P-producing block (glucose transporter and hexokinase) also showed high flux control (70%), which indicated low flux control by PFK-1. Kinetic analysis of PFK-1 showed low sensitivity towards its allosteric inhibitors citrate and ATP, at physiological concentrations of the activator Fru(2,6)P2. On the other hand, hexokinase activity was strongly inhibited by high, but physiological, concentrations of Glc6P. Therefore, the enhanced glycolytic flux in fast-growth tumor cells was still controlled by an over-produced, but Glc6P-inhibited hexokinase.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cell Proliferation , Glucose-6-Phosphate/chemistry , Hexokinase/antagonists & inhibitors , Hexokinase/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Female , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glycolysis , HeLa Cells , Hepatocytes/enzymology , Hepatocytes/metabolism , Hepatocytes/pathology , Hexokinase/biosynthesis , Hexokinase/chemistry , Humans , Kinetics , Lactic Acid/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Phosphofructokinase-1/metabolism , Rats , Rats, Wistar , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL