Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(8): 107500, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38944124

ABSTRACT

In eukaryotes, the D-enantiomer of arabinose (D-Ara) is an intermediate in the biosynthesis of D-erythroascorbate in yeast and fungi and in the biosynthesis of the nucleotide sugar GDP-α-D-arabinopyranose (GDP-D-Arap) and complex α-D-Arap-containing surface glycoconjugates in certain trypanosomatid parasites. Whereas the biosynthesis of D-Ara in prokaryotes is well understood, the route from D-glucose (D-Glc) to D-Ara in eukaryotes is unknown. In this paper, we study the conversion of D-Glc to D-Ara in the trypanosomatid Crithidia fasciculata using positionally labeled [13C]-D-Glc and [13C]-D-ribose ([13C]-D-Rib) precursors and a novel derivatization and gas chromatography-mass spectrometry procedure applied to a terminal metabolite, lipoarabinogalactan. These data implicate the both arms of pentose phosphate pathway and a likely role for D-ribulose-5-phosphate (D-Ru-5P) isomerization to D-Ara-5P. We tested all C. fasciculata putative sugar and polyol phosphate isomerase genes for their ability to complement a D-Ara-5P isomerase-deficient mutant of Escherichia coli and found that one, the glutamine fructose-6-phosphate aminotransferase (GFAT) of glucosamine biosynthesis, was able to rescue the E. coli mutant. We also found that GFAT genes of other trypanosomatid parasites, and those of yeast and human origin, could complement the E. coli mutant. Finally, we demonstrated biochemically that recombinant human GFAT can isomerize D-Ru-5P to D-Ara5P. From these data, we postulate a general eukaryotic pathway from D-Glc to D-Ara and discuss its possible significance. With respect to C. fasciculata, we propose that D-Ara is used not only for the synthesis of GDP-D-Arap and complex surface glycoconjugates but also in the synthesis of D-erythroascorbate.

2.
Anal Chem ; 96(4): 1774-1780, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38230524

ABSTRACT

In this paper, we demonstrate the existence of an endogenous mitochondrial azoreductase (AzoR) activity that can induce the cleavage of N═N double bonds of azobenzene compounds under normoxic conditions. To this end, 100% OFF-ON azo-based fluorogenic probes derived from 4-amino-1,8-naphthalimide fluorophores were synthesized and evaluated. The in vitro study conducted with other endogenous reducing agents of the cell, including reductases, demonstrated both the efficacy and the selectivity of the probe for AzoR. Confocal experiments with the probe revealed an AzoR activity in the mitochondria of living cells under normal oxygenation conditions, and we were able to demonstrate that this endogenous AzoR activity appears to be expressed at different levels across different cell lines. This discovery provides crucial information for our understanding of the biochemical processes occurring within the mitochondria. It thus contributes to a better understanding of its function, which is implicated in numerous pathologies.


Subject(s)
Amlodipine Besylate, Olmesartan Medoxomil Drug Combination , Naphthalimides , Nitroreductases , NADH, NADPH Oxidoreductases/metabolism , Fluorescent Dyes/chemistry
3.
Nat Commun ; 10(1): 1980, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040275

ABSTRACT

Here, we present a 3D localization-based super-resolution technique providing a slowly varying localization precision over a 1 µm range with precisions down to 15 nm. The axial localization is performed through a combination of point spread function (PSF) shaping and supercritical angle fluorescence (SAF), which yields absolute axial information. Using a dual-view scheme, the axial detection is decoupled from the lateral detection and optimized independently to provide a weakly anisotropic 3D resolution over the imaging range. This method can be readily implemented on most homemade PSF shaping setups and provides drift-free, tilt-insensitive and achromatic results. Its insensitivity to these unavoidable experimental biases is especially adapted for multicolor 3D super-resolution microscopy, as we demonstrate by imaging cell cytoskeleton, living bacteria membranes and axon periodic submembrane scaffolds. We further illustrate the interest of the technique for biological multicolor imaging over a several-µm range by direct merging of multiple acquisitions at different depths.


Subject(s)
Imaging, Three-Dimensional/methods , Microscopy/methods , Nanotechnology/methods , Algorithms , Anisotropy
4.
Article in English | MEDLINE | ID: mdl-29153997

ABSTRACT

The enzyme glucosamine-6-phosphate synthase (GlmS) is an important point of metabolic control in biosynthesis of amino sugar-containing macromolecules and is therefore a potential target in order to design antibacterial and antifungal drugs. It has two oligomerization states, which are the active dimer and the inactive hexamer. For the first time, the potential of CE to separate and quantify the two forms was studied. After incubating GlmS with the d-glucosamine 6-phosphate (GlcN6P) inhibitor, an electrolyte based on sodium phosphate at pH 7.2 and an ionic strength of 100mM plus GlcN6P (either 2 or 20mM) allowed the hexamer-dimer separation. However, the displacement of the dimer/hexamer equilibrium during the analysis time prevented any improvement of the resolution when varying the effective separation length or the temperature of the analysis. Therefore, the use of a short-end CE method allowed the decrease in the analysis time to about 1min. Some parameters such as the temperature and the time of incubation and the ratio of the inhibitor and enzyme concentrations were studied. Then, it was also possible to test, very rapidly and with a very small amount, some molecules having an inhibition potential for the GlmS enzyme (arabinose-5-phosphate oxime, 2-amino-2-deoxy-d-glucitol 6-phosphate, and glucose-6-phosphate).


Subject(s)
Electrophoresis, Capillary/methods , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/analysis , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Enzyme Inhibitors/analysis , Enzyme Inhibitors/metabolism , Escherichia coli Proteins/analysis , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/antagonists & inhibitors
5.
Nat Chem Biol ; 12(10): 787-794, 2016 10.
Article in English | MEDLINE | ID: mdl-27479744

ABSTRACT

Strigolactone plant hormones control plant architecture and are key players in both symbiotic and parasitic interactions. They contain an ABC tricyclic lactone connected to a butenolide group, the D ring. The DWARF14 (D14) strigolactone receptor belongs to the superfamily of α/ß-hydrolases, and is known to hydrolyze the bond between the ABC lactone and the D ring. Here we characterized the binding and catalytic functions of RAMOSUS3 (RMS3), the pea (Pisum sativum) ortholog of rice (Oryza sativa) D14 strigolactone receptor. Using new profluorescent probes with strigolactone-like bioactivity, we found that RMS3 acts as a single-turnover enzyme that explains its apparent low enzymatic rate. We demonstrated the formation of a covalent RMS3-D-ring complex, essential for bioactivity, in which the D ring was attached to histidine 247 of the catalytic triad. These results reveal an undescribed mechanism of plant hormone reception in which the receptor performs an irreversible enzymatic reaction to generate its own ligand.


Subject(s)
4-Butyrolactone/analogs & derivatives , Heterocyclic Compounds, 3-Ring/metabolism , Histidine/metabolism , Lactones/metabolism , Pisum sativum/metabolism , Plant Growth Regulators/metabolism , Plant Proteins/metabolism , 4-Butyrolactone/chemistry , 4-Butyrolactone/metabolism , Histidine/chemistry , Ligands , Molecular Structure , Pisum sativum/enzymology , Plant Growth Regulators/chemistry , Plant Proteins/chemistry
6.
Anal Biochem ; 458: 61-5, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24814295

ABSTRACT

Glucosamine-6-phosphate synthase (GlmS, EC 2.6.1.16) catalyzes the first and rate-limiting step in the hexosamine biosynthetic pathway, leading to the synthesis of uridine-5'-diphospho-N-acetyl-D-glucosamine, the major building block for the edification of peptidoglycan in bacteria, chitin in fungi, and glycoproteins in mammals. This bisubstrate enzyme converts D-fructose-6-phosphate (Fru-6P) and L-glutamine (Gln) into D-glucosamine-6-phosphate (GlcN-6P) and L-glutamate (Glu), respectively. We previously demonstrated that matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) allows determination of the kinetic parameters of the synthase activity. We propose here to refine the experimental protocol to quantify Glu and GlcN-6P, allowing determination of both hemisynthase and synthase parameters from a single assay kinetic experiment, while avoiding interferences encountered in other assays. It is the first time that MALDI-MS is used to survey the activity of a bisubstrate enzyme.


Subject(s)
Enzyme Assays , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Fructosephosphates/analysis , Fructosephosphates/metabolism , Glucosamine/analogs & derivatives , Glucosamine/analysis , Glucosamine/metabolism , Glucose-6-Phosphate/analogs & derivatives , Glucose-6-Phosphate/analysis , Glucose-6-Phosphate/metabolism , Glutamic Acid/analysis , Glutamic Acid/metabolism , Glutamine/analysis , Glutamine/metabolism , Kinetics , Substrate Specificity
7.
Biochimie ; 97: 39-48, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24075873

ABSTRACT

The enzyme glucosamine-6P Synthase (Gfat, L-glutamine:D-fructose-6P amidotransferase) is involved in the hexosamine biosynthetic pathway and catalyzes the formation of glucosamine-6P from the substrates d-fructose-6-phosphate and l-glutamine. In eukaryotic cells, Gfat is inhibited by UDPGlcNAc, the end product of the biochemical pathway. In this work we present the dissection of the binding and inhibition properties of this feedback inhibitor and of its fragments by a combination of STD-NMR experiments and inhibition measurements on the wild type human enzyme (hGfat) as well as on site-directed mutants. We demonstrate that the UDPGlcNAc binding site is located in the isomerase domain of hGfat. Two amino acid residues (G445 and G461) located at the bottom of the binding site are identified to play a key role in the specificity of UDPGlcNAc inhibition of hGfat activity vs its bacterial Escherichia coli counterpart. We also show that UDPGlcNAc subcomponents have distinct features: the nucleotidic moiety is entirely responsible for binding whereas the N-acetyl group is mandatory for inhibition but not for binding, and the sugar moiety acts as a linker between the nucleotidic and N-acetyl groups. Combining these structural recognition determinants therefore appears as a promising strategy to selectively inhibit hGfat, which may for example help reduce complications in diabetes.


Subject(s)
Fructosephosphates/metabolism , Glucosamine/analogs & derivatives , Glucose-6-Phosphate/analogs & derivatives , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Glutamine/metabolism , Uridine Diphosphate N-Acetylglucosamine/metabolism , Catalytic Domain , Escherichia coli/enzymology , Escherichia coli/genetics , Feedback, Physiological , Fructosephosphates/chemistry , Gene Expression , Glucosamine/chemistry , Glucosamine/metabolism , Glucose-6-Phosphate/chemistry , Glucose-6-Phosphate/metabolism , Glutamine/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Uridine Diphosphate N-Acetylglucosamine/chemistry
8.
J Biol Chem ; 287(41): 34533-46, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22851174

ABSTRACT

The amino-terminal cysteine of glucosamine-6-phosphate synthase (GlmS) acts as a nucleophile to release and transfer ammonia from glutamine to fructose 6-phosphate through a channel. The crystal structure of the C1A mutant of Escherichia coli GlmS, solved at 2.5 Å resolution, is organized as a hexamer, where the glutaminase domains adopt an inactive conformation. Although the wild-type enzyme is active as a dimer, size exclusion chromatography, dynamic and quasi-elastic light scattering, native polyacrylamide gel electrophoresis, and ultracentrifugation data show that the dimer is in equilibrium with a hexameric state, in vitro and in cellulo. The previously determined structures of the wild-type enzyme, alone or in complex with glucosamine 6-phosphate, are also consistent with a hexameric assembly that is catalytically inactive because the ammonia channel is not formed. The shift of the equilibrium toward the hexameric form in the presence of cyclic glucosamine 6-phosphate, together with the decrease of the specific activity with increasing enzyme concentration, strongly supports product inhibition through hexamer stabilization. Altogether, our data allow us to propose a morpheein model, in which the active dimer can rearrange into a transiently stable form, which has the propensity to form an inactive hexamer. This would account for a physiologically relevant allosteric regulation of E. coli GlmS. Finally, in addition to cyclic glucose 6-phosphate bound at the active site, the hexameric organization of E. coli GlmS enables the binding of another linear sugar molecule. Targeting this sugar-binding site to stabilize the inactive hexameric state is therefore suggested for the development of specific antibacterial inhibitors.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Models, Molecular , Protein Multimerization , Allosteric Regulation , Enzyme Stability , Escherichia coli Proteins/genetics , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Protein Structure, Quaternary , Structure-Activity Relationship
9.
J Am Soc Mass Spectrom ; 22(3): 431-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21472562

ABSTRACT

Escherichia coli glucosamine-6-phosphate synthase (GlmS) is a dimeric enzyme from the glutamine-dependent amidotransferases family, which catalyses the conversion of D-fructose-6-phosphate (Fru6P) and glutamine (Gln) into D-glucosamine-6-phosphate (GlcN6P) and glutamate, respectively. Extensive X-ray crystallography investigations have been reported, highlighting the importance of the dimeric association to form the sugar active site as well as significant conformational changes of the protein upon substrate and product binding. In the present work, an approach based on time-resolved noncovalent mass spectrometry has been developed to study the dynamics of GlmS subunit exchange. Using (14)N versus (15)N labeled proteins, the kinetics of GlmS subunit exchange was monitored with the wild-type enzyme in the presence of different substrates and products as well as with the protein bearing a key amino acid mutation specially designed to weaken the dimer interface. Determination of rate constants of subunit exchange revealed important modifications of the protein dynamics: while glutamine, glutamate, and K603A mutation accelerates subunit exchange, Fru6P and GlcN6P totally prevent it. These results are described in light of the available structural information, providing additional useful data for both the characterization of GlmS catalytic process and the design of new GlmS inhibitors. Finally, time-resolved noncovalent MS can be proposed as an additional biophysical technique for real-time monitoring of protein dynamics.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Amino Acid Substitution , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Glutamine/chemistry , Glutamine/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Nitrogen Isotopes , Protein Multimerization
10.
Arch Biochem Biophys ; 505(1): 1-12, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20709015

ABSTRACT

Glucosamine-6P synthase, which catalyzes glucosamine-6P synthesis from fructose-6P and glutamine, channels ammonia over 18Å between its glutaminase and synthase active sites. The crystal structures of the full-length Escherichia coli enzyme have been determined alone, in complex with the first bound substrate, fructose-6P, in the presence of fructose-6P and a glutamine analog, and in the presence of the glucosamine-6P product. These structures represent snapshots of reaction intermediates, and their comparison sheds light on the dynamics of catalysis. Upon fructose-6P binding, the C-terminal loop and the glutaminase domains get ordered, leading to the closure of the synthase site, the opening of the sugar ring and the formation of a "closed" ammonia channel. Then, glutamine binding leads to the closure of the Q-loop to protect the glutaminase site, the activation of the catalytic residues involved in glutamine hydrolysis, the swing of the side chain of Trp74, which allows the communication between the two active sites through an "open" channel, and the rotation of the glutaminase domains relative to the synthase domains dimer. Therefore, binding of the substrates at the appropriate reaction time is responsible for the formation and opening of the ammonia channel and for the activation of the enzyme glutaminase function.


Subject(s)
Escherichia coli/enzymology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Conformation
11.
Arch Biochem Biophys ; 498(2): 95-104, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20416269

ABSTRACT

Glucosamine-6-phosphate synthase (GlmS) is responsible for the first and rate-limiting step in the hexosamine biosynthetic pathway. It catalyzes the conversion of D-fructose-6P (F6P) into D-glucosamine-6P (GlcN6P) using L-glutamine (Gln) as nitrogen donor (synthase activity) according to an ordered bi-bi process where F6P binds first. In the absence of F6P, the enzyme exhibits a weak hydrolyzing activity of Gln into Glu and ammonia (glutaminase activity), whereas the presence of F6P strongly stimulates it (hemi-synthase activity). Until now, these different activities were indirectly measured using either coupled enzyme or colorimetric methods. In this work, we have developed a direct assay monitoring the heat released by the reaction. Isothermal titration calorimetry and differential scanning calorimetry were used to determine kinetic and thermodynamic parameters of GlmS. The direct determination at 37 degrees C of kinetic parameters and affinity constants for both F6P and Gln demonstrated that part of the ammonia produced by Gln hydrolysis in the presence of both substrates is not used for the formation of the GlcN6P. The full characterization of this phenomenon allowed to identify experimental conditions where this leak of ammonia is negligible. Enthalpy measurements at 25 degrees C in buffers of various heats of protonation demonstrated that no proton exchange with the medium occurred during the enzyme-catalyzed glutaminase or synthase reaction suggesting for the first time that both products are released as a globally neutral pair composed by the Glu carboxylic side chain and the GlcN6P amine function. Finally we showed that the oligomerization state of GlmS is concentration-dependent.


Subject(s)
Escherichia coli/enzymology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Calorimetry, Differential Scanning , Calorimetry, Indirect , Catalysis , Escherichia coli Proteins , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Glucose-6-Phosphate/analogs & derivatives , Glucose-6-Phosphate/chemistry , Hot Temperature , Kinetics
12.
J Mol Biol ; 385(2): 653-64, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-18976669

ABSTRACT

The large protein motions of the bacterial enzyme glucosamine-6-phosphate synthase have been addressed using full atom normal modes analysis for the empty, the glucose-6-phosphate and the glucose-6-phosphate+glutamate bound proteins. The approach that was used involving energy minimizations along the normal modes coordinates identified functional motions of the protein, some of which were characterized earlier by X-ray diffraction studies. This method made it possible for the first time to highlight significant energy differences according to whether none, only one or both of the active sites of the protein were occupied. Our data favoured a specific motion of the glutamine binding domain following the fixation of fructose-6-phosphate and suggested a rigidified structure with both sites occupied. Here, we show that most of the collective large amplitude motions of glucosamine-6-phosphate synthase that are modulated by ligand binding are crucial for the enzyme catalytic cycle, as they strongly modify the geometry of both the ammonia channel and the C-tail, demonstrating their role in ammonia transfer and ligand binding.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Motion , Ammonia/metabolism , Binding Sites , Crystallography, X-Ray , Fructosephosphates/metabolism , Ligands , Models, Molecular , Protein Structure, Tertiary
13.
J Mol Biol ; 377(4): 1174-85, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18295797

ABSTRACT

Glucosamine-6-phosphate synthase (GlmS) channels ammonia from glutamine at the glutaminase site to fructose 6-phosphate (Fru6P) at the synthase site. Escherichia coli GlmS is composed of two C-terminal synthase domains that form the dimer interface and two N-terminal glutaminase domains at its periphery. We report the crystal structures of GlmS alone and in complex with the glucosamine-6-phosphate product at 2.95 A and 2.9 A resolution, respectively. Surprisingly, although the whole protein is present in this crystal form, no electron density for the glutaminase domain was observed, indicating its mobility. Comparison of the two structures with that of the previously reported GlmS-Fru6P complex shows that, upon sugar binding, the C-terminal loop, which forms the major part of the channel walls, becomes ordered and covers the synthase site. The ordering of the glutaminase domains likely follows Fru6P binding by the anchoring of Trp74, which acts as the gate of the channel, on the closed C-terminal loop. This is accompanied by a major conformational change of the side chain of Lys503# of the neighboring synthase domain that strengthens the interactions of the synthase domain with the C-terminal loop and completely shields the synthase site. The concomitant conformational change of the Lys503#-Gly505# tripeptide places catalytic His504# in the proper position to open the sugar and buries the linear sugar, which is now in the vicinity of the catalytic groups involved in the sugar isomerization reaction. Together with the previously reported structures of GlmS in complex with Fru6P or glucose 6-phosphate and a glutamine analogue, the new structures reveal the structural changes occurring during the whole catalytic cycle.


Subject(s)
Ammonia/metabolism , Glucosamine/analogs & derivatives , Glucose-6-Phosphate/analogs & derivatives , Glutaminase/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/physiology , Protein Folding , Carbohydrates/chemistry , Catalytic Domain/drug effects , Crystallography, X-Ray , Cyclization , Escherichia coli/enzymology , Glucosamine/chemistry , Glucosamine/metabolism , Glucose-6-Phosphate/chemistry , Glucose-6-Phosphate/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Models, Molecular , Protein Structure, Tertiary/physiology , Solvents/pharmacology
14.
Arch Biochem Biophys ; 474(2): 302-17, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18279655

ABSTRACT

L-Glutamine:d-fructose-6-phosphate amidotransferase, also known as glucosamine-6-phosphate synthase (GlcN6P synthase), which catalyzes the first step in a pathway leading to the formation of uridine 5'-diphospho-N-acetyl-d-glucosamine (UDP-GlcNAc), is a key point in the metabolic control of the biosynthesis of amino sugar-containing macromolecules. The molecular mechanism of the reaction catalyzed by GlcN6P synthase is complex and involves amide bond cleavage followed by ammonia channeling and sugar isomerization. This article provides a comprehensive overview of the present knowledge on this multi-faceted enzyme emphasizing the progress made during the last five years.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Models, Molecular , Uridine Diphosphate N-Acetylglucosamine/metabolism , Ammonia/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Candida albicans/enzymology , Catalysis , Enzyme Activation , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Gene Expression Regulation, Enzymologic , Glucose/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/biosynthesis , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Humans , Isomerism , Kinetics , Structure-Activity Relationship , Thermodynamics
15.
Biochemistry ; 46(45): 13163-9, 2007 Nov 13.
Article in English | MEDLINE | ID: mdl-17941647

ABSTRACT

Glutamine:fructose-6-phosphate amidotransferase (Gfat) catalyzes the first and rate-limiting step in the hexosamine biosynthetic pathway. The increasing amount of evidence that links excess hexosamine biosynthesis with pathogenic complications of type II diabetes highlights the need to understand the regulation of Gfat. Previous studies showed that eukaryotic Gfat is subjected to feedback inhibition by UDP-N-acetyl-d-glucosamine (UDP-GlcNAc) and to phosphorylation by cAMP-activated protein kinase A (PKA). In this study, overexpression of human Gfat isoform 1 (hGfat1) in insect cells revealed that hGfat1 is phosphorylated in vivo. Using matrix-assisted laser desorption/ionization and electrospray tandem mass spectrometry, we have identified Ser243 as a novel phosphorylation site. Biochemical properties of the wild type and the Ser243Glu mutant of hGfat1 overexpressed in Escherichia coli were compared. Our results provide evidence that phosphorylation at Ser243 stimulates glucosamine 6-phosphate-synthesizing activity, lowers amidohydrolyzing activity in the absence of fructose 6-phosphate (F6P) (glutaminase activity), and lowers Km(F6P) 2-fold, but has no effect on UDP-GlcNAc inhibition. On the basis of the sequence consensus, AMP-activated protein kinase and calcium/calmodulin-dependent kinase II were identified to phosphorylate specifically Ser243 in vitro. Phosphorylation by these two kinases results in an increase of enzymatic activity by 1.4-fold. These findings suggest for the first time that hGfat1 may be regulated by kinases other than PKA.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Serine/chemistry , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Phosphorylation , Sequence Alignment , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spodoptera , Tandem Mass Spectrometry
16.
Anal Biochem ; 370(2): 142-6, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17880906

ABSTRACT

An assay for glucosamine-6-phosphate synthase using a yeast glucosamine-6-phosphate N-acetyltransferase 1 (GNA1) as coupling enzyme was developed. GNA1 transfers the acetyl moiety from acetyl-coenzyme A (CoA) to glucosamine-6-phosphate, releasing coenzyme A. The assay measures the production of glucosamine-6-phosphate by either following the consumption of acetyl-CoA spectrophotometrically at 230nm or quantifying the free thiol with 5,5'-dithio-bis(2-nitrobenzoic acid) (Ellman's reagent) in a discontinuous manner. This method is simple to perform and can be adapted to a 96-well microtiter plate format, which will facilitate high-throughput inhibitor screening and mechanistic studies using purified GlmS.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Transaminases/metabolism , Kinetics , Polymerase Chain Reaction , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
17.
FEBS Lett ; 581(16): 2981-7, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17559838

ABSTRACT

Ammonia transfer from the glutamine site to the fructose-6P site of bacterial glucosamine-6-phosphate synthase was studied by molecular dynamics simulations. The studies suggest a key role for Trp74, in the sealing of the hydrophobic channel connecting the two binding sites, as well as for the two Ala602 and Val605 residues, which form a narrow passage whose opening/closing constitutes an essential event in ammonia transfer. Kinetic analyses of the corresponding protein mutants confirmed our predictions. The efficiency of ammonia transfer which was close to zero in the W74A mutant was partially restored by increasing the size of the corresponding side-chain; the simulations performed on the W74A mutant suggested the formation of a hole in the channel. In the case of A602L and V605L mutants, the efficiency of ammonia transfer decreased to approximately 50% of the value of the native protein. None of the mutants were, however, able to use exogenous ammonia as a substrate.


Subject(s)
Ammonia/metabolism , Computer Simulation , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Ammonia/chemistry , Bacterial Proteins/metabolism , Binding Sites , Biological Transport , Crystallography, X-Ray , Escherichia coli/genetics , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Glucosamine/metabolism , Glucose-6-Phosphate/analogs & derivatives , Glucose-6-Phosphate/chemistry , Glucose-6-Phosphate/metabolism , Glutamine/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Hydrolysis , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/metabolism , Point Mutation
18.
Protein Expr Purif ; 54(1): 45-53, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17379537

ABSTRACT

Human L-glutamine: D-fructose-6-phosphate amidotransferase (Gfat1), a recognized target in type 2 diabetes complications, was expressed in Sf9 insect cells with an internal His(6)-tag and purified to homogenity. Two different microplate assays that quantify, respectively D-glucosamine-6-phosphate and L-glutamate were used to analyze the enzyme kinetic properties. The recombinant human L-glutamine: D-fructose-6-phosphate amidotransferase isoform 1 exhibits Michaelis parameters K(m)(Fru-6P)=0.98 mM and K(m)(Gln)=0.84 mM which are similar to the values reported for the same enzyme from different sources. The stimulation of hydrolysis of the alternate substrate L-glutamine para-nitroanilide by D-fructose-6P (Fru-6P) afforded a K(d) of 5 microM for Fru-6P.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/biosynthesis , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/isolation & purification , Histidine/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Animals , Cells, Cultured , Circular Dichroism , Cloning, Molecular , Enzyme Activation , Fructosephosphates/chemistry , Glutamic Acid/chemistry , Glutaminase/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Humans , Hydrolysis , Insecta/cytology , Insecta/metabolism , Kinetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry
19.
Bioorg Med Chem Lett ; 17(7): 1966-70, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17291748

ABSTRACT

Results of an in silico screening of a freely accessible database encompassing 50,000 commercial compounds on bacterial glucosamine-6P synthase (Glms) are described. Each product was docked with the GOLD software in a region of 20A surrounding the sugar binding site and ranked according to its score. Among the 14 best-scored molecules, three molecules exhibited good experimental inhibition properties (IC(50)=70 microM) giving a high hit rate (H.R.: 0.23). Interestingly, these molecules are predicted to interact with a protein region that forms a pocket at the interface between the two enzyme monomers, opening the route to dimerization inhibitors.


Subject(s)
Chemistry, Pharmaceutical/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Binding Sites , Carbohydrates/chemistry , Computer Simulation , Dimerization , Drug Design , Enzyme Inhibitors/chemical synthesis , Inhibitory Concentration 50 , Kinetics , Ligands , Models, Chemical , Molecular Conformation , Protein Binding , Software
20.
FEBS Lett ; 580(22): 5130-6, 2006 Oct 02.
Article in English | MEDLINE | ID: mdl-16962102

ABSTRACT

We demonstrate the utility of normal mode analysis in correctly predicting the binding modes of inhibitors in the active sites of matrix metalloproteinases (MMPs). We show the accuracy in predicting the positions of MMP-3 inhibitors is strongly dependent on which structure is used as the target, especially when it has been energy minimized. This dependency can be overcome by using intermediate structures generated along one of the normal modes previously calculated for a given target. These results may be of prime importance for further in silico drug discovery.


Subject(s)
Drug Design , Matrix Metalloproteinase 3/chemistry , Models, Chemical , Models, Molecular , Protease Inhibitors/chemistry , Software , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...