Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Bioorg Med Chem Lett ; 102: 129666, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38382679

RESUMEN

Because tuberculosis is still a major health threat worldwide, identification of new drug targets is urgently needed. In this study, we considered type B ribose-5-phosphate isomerase from Mycobacterium tuberculosis as a potential target, and addressed known problems of previous inhibitors in terms of their sensitivity to hydrolysis catalyzed by phosphatase enzymes, which impaired their potential use as drugs. To this end, we synthesized six novel phosphomimetic compounds designed to be hydrolytically stable analogs of the substrate ribose 5-phosphate and the best known inhibitor 5-phospho-d-ribonate. The phosphate function was replaced by phosphonomethyl, sulfate, sulfonomethyl, or malonate groups. Inhibition was evaluated on type A and type B ribose-5-phosphate isomerases, and stability towards hydrolysis using alkaline phosphatase and veal serum was assessed. One of the phosphomimetic analogs, 5-deoxy-5-phosphonomethyl-d-ribonate, emerged as the first strong and specific inhibitor of the M. tuberculosis enzyme that is resistant to hydrolysis.

2.
J Comput Chem ; 41(8): 839-854, 2020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-31909840

RESUMEN

We calibrate and validate the parameters necessary to represent the dianionic phosphate group (DPG) in molecular mechanics. DPG is an essential fragment of signaling biological molecules and protein-binding ligands. It is a constitutive fragment of biosensors, which bind to the dimer interface of phosphoglucose isomerase (PGI), an intracellular enzyme involved in sugar metabolism, as well as an extracellular protein known as autocrine motility factor (AMF) closely related to metastasis formation. Our long-term objective is to design DPG-based biosensors with enhanced affinities for AMF/PGI cancer biomarker in blood. Molecular dynamics with polarizable potentials could be used toward this aim. This requires to first evaluate the accuracy of such potentials upon representing the interactions of DPG with its PGI ligands and tightly bound water molecules. Such evaluations are done by comparisons with high-level ab initio quantum chemistry (QC) calculations. We focus on the Sum of Interactions Between Fragments Ab initio computed (SIBFA) polarizable molecular mechanics procedure. We present first the results of the DPG calibration. This is followed by comparisons between ΔE(SIBFA) and ΔE(QC) regarding bi-molecular complexes of DPG with the main-chain and side-chain PGI residues, which bind to it in the recognition site. We then consider DPG complexes with an increasing number of PGI residues. The largest QC complexes encompass the entirety of the recognition site, with six structural water molecules totaling up to 211 atoms. A persistent and satisfactory agreement could be shown between ΔE(SIBFA) and ΔE(QC). These validations constitute an essential first step toward large-scale molecular dynamics simulations of DPG-based biosensors bound at the PGI dimer interface. © 2020 Wiley Periodicals, Inc.


Asunto(s)
Teoría Funcional de la Densidad , Glucosa-6-Fosfato Isomerasa/química , Fosfatos/química , Aniones/química , Calibración , Glucosa-6-Fosfato Isomerasa/metabolismo
3.
Bioorg Chem ; 102: 104048, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32682158

RESUMEN

Phosphoglucose isomerase (PGI) is a cytosolic enzyme that catalyzes the reversible interconversion of d-glucose 6-phosphate and d-fructose 6-phosphate in glycolysis. Outside the cell, PGI is also known as autocrine motility factor (AMF), a cytokine secreted by a large variety of tumor cells that stimulates motility of cancer cells in vitro and metastases development in vivo. Human PGI and AMF are strictly identical proteins both in terms of sequence and 3D structure, and AMF activity is known to involve, at least in part, the enzymatic active site. Hence, with the purpose of finding new strong AMF-PGI inhibitors that could be potentially used as anticancer agents and/or as bioreceptors for carbohydrate-based electrochemical biosensors, we report in this study the synthesis and kinetic evaluation of several new human PGI inhibitors derived from the synthon 5-phospho-d-arabinono-1,4-lactone. Although not designed as high-energy intermediate analogue inhibitors of the enzyme catalyzed isomerization reaction, several of these N-substituted 5-phosphate-d-arabinonamide derivatives appears as new strong PGI inhibitors. For one of them, we report its crystal structure in complex with human PGI at 2.38 Å. Detailed analysis of its interactions at the active site reveals a new binding mode and shows that human PGI is relatively tolerant for modified inhibitors at the "head" C-1 part, offering promising perspectives for the future design of carbohydrate-based biosensors.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Glucosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Fosfatos/síntesis química , Fosfatos/uso terapéutico , Inhibidores Enzimáticos/farmacología , Humanos , Fosfatos/farmacología , Relación Estructura-Actividad
4.
J Biol Chem ; 290(18): 11293-308, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25792735

RESUMEN

The riboflavin biosynthesis pathway has been shown to be essential in many pathogens and is absent in humans. Therefore, enzymes involved in riboflavin synthesis are considered as potential antibacterial drug targets. The enzyme 3,4-dihydroxy-2-butanone-4-phosphate synthase (DHBPS) catalyzes one of the two committed steps in the riboflavin pathway and converts d-ribulose 5-phosphate (Ru5P) to l-3,4-dihydroxy-2-butanone 4-phosphate and formate. Moreover, DHBPS is shown to be indispensable for Mycobacterium, Salmonella, and Helicobacter species. Despite the essentiality of this enzyme in bacteria, no inhibitor has been identified hitherto. Here, we describe kinetic and crystal structure characterization of DHBPS from Vibrio cholerae (vDHBPS) with a competitive inhibitor 4-phospho-d-erythronohydroxamic acid (4PEH) at 1.86-Å resolution. In addition, we also report the structural characterization of vDHBPS in its apo form and in complex with its substrate and substrate plus metal ions at 1.96-, 1.59-, and 2.04-Å resolution, respectively. Comparison of these crystal structures suggests that 4PEH inhibits the catalytic activity of DHBPS as it is unable to form a proposed intermediate that is crucial for DHBPS activity. Furthermore, vDHBPS structures complexed with substrate and metal ions reveal that, unlike Candida albicans, binding of substrate to vDHBPS induces a conformational change from an open to closed conformation. Interestingly, the position of second metal ion, which is different from that of Methanococcus jannaschii, strongly supports an active role in the catalytic mechanism. Thus, the kinetic and structural characterization of vDHBPS reveals the molecular mechanism of inhibition shown by 4PEH and that it can be explored further for designing novel antibiotics.


Asunto(s)
Unión Competitiva , Inhibidores Enzimáticos/farmacología , Ácidos Hidroxámicos/farmacología , Ligasas/antagonistas & inhibidores , Ligasas/química , Fosfatos de Azúcar/metabolismo , Fosfatos de Azúcar/farmacología , Vibrio cholerae/enzimología , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/metabolismo , Ácidos Hidroxámicos/metabolismo , Cinética , Ligasas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Riboflavina/biosíntesis
5.
J Comput Chem ; 37(32): 2770-2782, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27699809

RESUMEN

Zn-metalloproteins are a major class of targets for drug design. They constitute a demanding testing ground for polarizable molecular mechanics/dynamics aimed at extending the realm of quantum chemistry (QC) to very long-duration molecular dynamics (MD). The reliability of such procedures needs to be demonstrated upon comparing the relative stabilities of competing candidate complexes of inhibitors with the recognition site stabilized in the course of MD. This could be necessary when no information is available regarding the experimental structure of the inhibitor-protein complex. Thus, this study bears on the phosphomannose isomerase (PMI) enzyme, considered as a potential therapeutic target for the treatment of several bacterial and parasitic diseases. We consider its complexes with 5-phospho-d-arabinonohydroxamate and three analog ligands differing by the number and location of their hydroxyl groups. We evaluate the energy accuracy expectable from a polarizable molecular mechanics procedure, SIBFA. This is done by comparisons with ab initio quantum-chemistry (QC) calculations in the following cases: (a) the complexes of the four ligands in three distinct structures extracted from the entire PMI-ligand energy-minimized structures, and totaling up to 264 atoms; (b) the solvation energies of several energy-minimized complexes of each ligand with a shell of 64 water molecules; (c) the conformational energy differences of each ligand in different conformations characterized in the course of energy-minimizations; and (d) the continuum solvation energies of the ligands in different conformations. The agreements with the QC results appear convincing. On these bases, we discuss the prospects of applying the procedure to ligand-macromolecule recognition problems. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Ácidos Hidroxámicos/química , Metaloproteínas/química , Simulación de Dinámica Molecular , Teoría Cuántica , Fosfatos de Azúcar/química , Zinc/química , Sitios de Unión , Ligandos , Metaloproteínas/metabolismo , Zinc/metabolismo
6.
Biochem J ; 449(1): 175-87, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23039077

RESUMEN

Malaria tropica is a devastating infectious disease caused by Plasmodium falciparum. This parasite synthesizes vitamin B6 de novo via the PLP (pyridoxal 5'-phosphate) synthase enzymatic complex consisting of PfPdx1 and PfPdx2 proteins. Biosynthesis of PLP is largely performed by PfPdx1, ammonia provided by PfPdx2 subunits is condensed together with R5P (D-ribose 5-phosphate) and G3P (DL-glyceraldehyde 3-phosphate). PfPdx1 accommodates both the R5P and G3P substrates and intricately co-ordinates the reaction mechanism, which is composed of a series of imine bond formations, leading to the production of PLP. We demonstrate that E4P (D-erythrose 4-phosphate) inhibits PfPdx1 in a dose-dependent manner. We propose that the acyclic phospho-sugar E4P, with a C1 aldehyde group similar to acyclic R5P, could interfere with R5P imine bond formations in the PfPdx1 reaction mechanism. Molecular docking and subsequent screening identified the E4P hydrazide analogue 4PEHz (4-phospho-D-erythronhydrazide), which selectively inhibited PfPdx1 with an IC50 of 43 µM. PfPdx1 contained in the heteromeric PLP synthase complex was shown to be more sensitive to 4PEHz and was inhibited with an IC50 of 16 µM. Moreover, the compound had an IC50 value of 10 µM against cultured P. falciparum intraerythrocytic parasites. To analyse further the selectivity of 4PEHz, transgenic cell lines overexpressing PfPdx1 and PfPdx2 showed that additional copies of the protein complex conferred protection against 4PEHz, indicating that the PLP synthase is directly affected by 4PEHz in vivo. These PfPdx1 inhibitors represent novel lead scaffolds which are capable of targeting PLP biosynthesis, and we propose this as a viable strategy for the development of new therapeutics against malaria.


Asunto(s)
Antimaláricos/farmacología , Plasmodium falciparum/efectos de los fármacos , Complejo Piruvato Deshidrogenasa/antagonistas & inhibidores , Animales , Antimaláricos/química , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Plasmodium falciparum/fisiología , Complejo Piruvato Deshidrogenasa/química , Especificidad por Sustrato , Fosfatos de Azúcar/química , Fosfatos de Azúcar/farmacología
7.
Bioorg Med Chem ; 20(4): 1511-20, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22269276

RESUMEN

In the design of inhibitors of phosphosugar metabolizing enzymes and receptors with therapeutic interest, malonate has been reported in a number of cases as a good and hydrolytically-stable surrogate of the phosphate group, since both functions are dianionic at physiological pH and of comparable size. We have investigated a series of malonate-based mimics of the best known phosphate inhibitors of class II (zinc) fructose-1,6-bis-phosphate aldolases (FBAs) (e.g., from Mycobacterium tuberculosis), type I (zinc) phosphomannose isomerase (PMI) from Escherichia coli, and phosphoglucose isomerase (PGI) from yeast. In the case of FBAs, replacement of one phosphate by one malonate on a bis-phosphorylated inhibitor (1) led to a new compound (4) still showing a strong inhibition (K(i) in the nM range) and class II versus class I selectivity (up to 8×10(4)). Replacement of the other phosphate however strongly affected binding efficiency and selectivity. In the case of PGI and PMI, 5-deoxy-5-malonate-D-arabinonohydroxamic acid (8) yielded a strong decrease in binding affinities when compared to its phosphorylated parent compound 5-phospho-D-arabinonohydroxamic acid (2). Analysis of the deposited 3D structures of the kinetically evaluated enzymes complexed to the phosphate-based inhibitors indicate that malonate could be a good phosphate surrogate only if phosphate is not tightly bound at the enzyme active site, such as in position 7 of compound 1 for FBAs. These observations are of importance for further design of inhibitors of phosphorylated-compounds metabolizing enzymes with therapeutic interest.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Fructosa-Bifosfato Aldolasa/antagonistas & inhibidores , Glucosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Malonatos/síntesis química , Manosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Animales , Dominio Catalítico , Activación Enzimática/efectos de los fármacos , Escherichia/enzimología , Humanos , Concentración 50 Inhibidora , Malonatos/química , Malonatos/farmacología , Modelos Biológicos , Estructura Molecular , Levaduras/enzimología
8.
Proteins ; 79(1): 203-20, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21058398

RESUMEN

Type I phosphomannose isomerases (PMIs) are zinc-dependent metalloenzymes involved in the reversible isomerization of D-mannose 6-phosphate (M6P) and D-fructose 6-phosphate (F6P). 5-Phospho-D-arabinonohydroxamic acid (5PAH), an inhibitor endowed with nanomolar affinity for yeast (Type I) and Pseudomonas aeruginosa (Type II) PMIs (Roux et al., Biochemistry 2004; 43:2926-2934), strongly inhibits human (Type I) PMI (for which we report an improved expression and purification procedure), as well as Escherichia coli (Type I) PMI. Its K(i) value of 41 nM for human PMI is the lowest value ever reported for an inhibitor of PMI. 5-Phospho-D-arabinonhydrazide, a neutral analogue of the reaction intermediate 1,2-cis-enediol, is about 15 times less efficient at inhibiting both enzymes, in accord with the anionic nature of the postulated high-energy reaction intermediate. Using the polarizable molecular mechanics, sum of interactions between fragments ab initio computed (SIBFA) procedure, computed structures of the complexes between Candida albicans (Type I) PMI and the cyclic substrate ß-D-mannopyranose 6-phosphate (ß-M6P) and between the enzyme and the high-energy intermediate analogue inhibitor 5PAH are reported. Their analysis allows us to identify clearly the nature of each individual active site amino acid and to formulate a hypothesis for the overall mechanism of the reaction catalyzed by Type I PMIs, that is, the ring-opening and isomerization steps, respectively. Following enzyme-catalyzed ring-opening of ß-M6P by zinc-coordinated water and Gln111 ligands, Lys136 is identified as the probable catalytic base involved in proton transfer between the two carbon atoms C1 and C2 of the substrate D-mannose 6-phosphate.


Asunto(s)
Manosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Manosa-6-Fosfato Isomerasa/química , Secuencia de Aminoácidos , Unión Competitiva , Candida albicans/enzimología , Dominio Catalítico , Escherichia coli/enzimología , Fructosafosfatos/química , Humanos , Hidrazinas/química , Ácidos Hidroxámicos/química , Cinética , Manosa-6-Fosfato Isomerasa/biosíntesis , Manosafosfatos/química , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Conformación Proteica , Alineación de Secuencia , Fosfatos de Azúcar/química
9.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 8): 934-44, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20693693

RESUMEN

Crystallographic binding studies have been carried out to probe the active-site binding properties of a monomeric variant (A-TIM) of triosephosphate isomerase (TIM). These binding studies are part of a structure-based directed-evolution project aimed towards changing the substrate specificity of monomeric TIM and are therefore aimed at finding binders which are substrate-like molecules. A-TIM has a modified more extended binding pocket between loop-7 and loop-8 compared with wild-type TIM. The A-TIM crystals were grown in the presence of citrate, which is bound in the active site of each of the two molecules in the asymmetric unit. In this complex, the active-site loops loop-6 and loop-7 adopt the closed conformation, similar to that observed in liganded wild-type TIM. Extensive crystal-soaking protocols have been developed to flush the bound citrate out of the active-site pocket of both molecules and the crystal structure shows that the unliganded open conformation of the A-TIM active site is the same as in unliganded wild-type TIM. It is also shown that sulfonate compounds corresponding to the transition-state analogue 2-phosphoglycolate bind in the active site, which has a closed conformation. It is also shown that the new binding pocket of A-TIM can bind 3-phosphoglycerate (3PGA; an analogue of a C4-sugar phosphate) and 4-phospho-D-erythronohydroxamic acid (4PEH; an analogue of a C5-sugar phosphate). Therefore, these studies have provided a rationale for starting directed-evolution experiments aimed at generating the catalytic properties of a C5-sugar phosphate isomerase on the A-TIM framework.


Asunto(s)
Dominios y Motivos de Interacción de Proteínas , Triosa-Fosfato Isomerasa/química , Trypanosoma brucei brucei/enzimología , Secuencia de Aminoácidos , Cristalografía por Rayos X , Modelos Moleculares , Datos de Secuencia Molecular , Ingeniería de Proteínas , Estructura Secundaria de Proteína
10.
Bioorg Med Chem ; 17(20): 7100-7, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19783448

RESUMEN

Non-hydrolyzable d-mannose 6-phosphate analogues in which the phosphate group was replaced by a phosphonomethyl, a dicarboxymethyl, or a carboxymethyl group were synthesized and kinetically evaluated as substrate analogues acting as potential inhibitors of type I phosphomannose isomerases (PMIs) from Saccharomyces cerevisiae and Escherichia coli. While 6-deoxy-6-phosphonomethyl-d-mannose and 6-deoxy-6-carboxymethyl-D-mannose did not inhibit the enzymes significantly, 6-deoxy-6-dicarboxymethyl-D-mannose appeared as a new strong competitive inhibitor of both S. cerevisiae and E. coli PMIs with K(m)/K(i) ratios of 28 and 8, respectively. We thus report the first malonate-based inhibitor of an aldose-ketose isomerase to date. Phosphonomethyl mimics of the 1,2-cis-enediolate high-energy intermediate postulated for the isomerization reaction catalyzed by PMIs were also synthesized but behave as poor inhibitors of PMIs. A polarizable molecular mechanics (SIBFA) study was performed on the complexes of d-mannose 6-phosphate and two of its analogues with PMI from Candida albicans, an enzyme involved in yeast infection homologous to S. cerevisiae and E. coli PMIs. It shows that effective binding to the catalytic site occurs with retention of the Zn(II)-bound water molecule. Thus the binding of the hydroxyl group on C1 of the ligand to Zn(II) should be water-mediated. The kinetic study reported here also suggests the dianionic character of the phosphate surrogate as a likely essential parameter for strong binding of the inhibitor to the enzyme active site.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Manosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Manosafosfatos/síntesis química , Manosafosfatos/farmacología , Ácidos Urónicos/farmacología , Cromatografía por Intercambio Iónico , Evaluación Preclínica de Medicamentos , Cinética , Espectroscopía de Resonancia Magnética , Manosa-6-Fosfato Isomerasa/química , Manosa-6-Fosfato Isomerasa/metabolismo , Modelos Moleculares , Saccharomyces cerevisiae/enzimología , Espectrometría de Masa por Ionización de Electrospray , Especificidad por Sustrato
11.
Appl Spectrosc ; 62(2): 213-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18284798

RESUMEN

The aim of this work was to perform highly localized spectroscopic surface measurements by combining time-resolved laser spectroscopy and scanning near-field optical microscopy. The final purpose of that was to study surface sorption at the molecular level of trivalent ions in the framework of nuclear waste disposal assessment. Time-resolved laser spectroscopy presents the advantages of being selective, sensitive, and noninvasive and scanning near-field optical microscopy is a promising technique for high resolution surface speciation. Investigation of the interaction between trivalent europium and a monocrystalline alumina (1102) surface was made using different conditions of concentration and pH. We found that the distribution of sorbed europium was always homogeneous with a decay time of europium (III) equal to 350 micros+/-15 micros. On the other hand, carbonate species with a decay time of 210 micros+/-10 micros or other hydroxide species with a decay time of 180 micros+/-10 micros were detected on the surface when a higher concentration or a higher pH solution, respectively, were used. Distribution of these species was heterogeneous and their associated fluorescence signal was relatively high, evoking a precipitated form. X-ray photoelectron spectroscopy (XPS) was also used on the same samples as a complementary technique. A binding energy of 1135.1 eV was obtained for the sorbed europium and another binding energy of 1134.4 eV was obtained for the hydroxide species, thus confirming the presence of two kinds of species on the surface.

12.
Biochem J ; 401(1): 279-85, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16981853

RESUMEN

Trypanosoma cruzi, the human parasite that causes Chagas disease, contains a functional pentose phosphate pathway, probably essential for protection against oxidative stress and also for R5P (ribose 5-phosphate) production for nucleotide synthesis. The haploid genome of the CL Brener clone of the parasite contains one gene coding for a Type B Rpi (ribose 5-phosphate isomerase), but genes encoding Type A Rpis, most frequent in eukaryotes, seem to be absent. The RpiB enzyme was expressed in Escherichia coli as a poly-His tagged active dimeric protein, which catalyses the reversible isomerization of R5P to Ru5P (ribulose 5-phosphate) with Km values of 4 mM (R5P) and 1.4 mM (Ru5P). 4-phospho-D-erythronohydroxamic acid, an analogue to the reaction intermediate when the Rpi acts via a mechanism involving the formation of a 1,2-cis-enediol, inhibited the enzyme competitively, with an IC50 value of 0.7 mM and a Ki of 1.2 mM. Site-directed mutagenesis allowed the demonstration of a role for His102, but not for His138, in the opening of the ribose furanosic ring. A major role in catalysis was confirmed for Cys69, since the C69A mutant was inactive in both forward and reverse directions of the reaction. The present paper contributes to the know-ledge of the mechanism of the Rpi reaction; in addition, the absence of RpiBs in the genomes of higher animals makes this enzyme a possible target for chemotherapy of Chagas disease.


Asunto(s)
Isomerasas Aldosa-Cetosa/metabolismo , Proteínas Protozoarias/metabolismo , Trypanosoma cruzi/enzimología , Isomerasas Aldosa-Cetosa/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Secuencia Conservada , Cinética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido
13.
J Med Chem ; 61(23): 10558-10572, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30418024

RESUMEN

The glycolytic enzyme aldolase is an emerging drug target in diseases such as cancer and protozoan infections which are dependent on a hyperglycolytic phenotype to synthesize adenosine 5'-triphosphate and metabolic precursors for biomass production. To date, structural information for the enzyme in complex with phosphate-derived inhibitors has been lacking. Thus, we determined the crystal structure of mammalian aldolase in complex with naphthalene 2,6-bisphosphate (1) that served as a template for the design of bisphosphonate-based inhibitors, namely, 2-phosphate-naphthalene 6-bisphosphonate (2), 2-naphthol 6-bisphosphonate (3), and 1-phosphate-benzene 4-bisphosphonate (4). All inhibitors targeted the active site, and the most promising lead, 2, exhibited slow-binding inhibition with an overall inhibition constant of ∼38 nM. Compound 2 inhibited proliferation of HeLa cancer cells, whereas HEK293 cells expressing a normal phenotype were not inhibited. The crystal structures delineated the essential features of high-affinity phosphate-derived inhibitors and provide a template for the development of inhibitors with prophylaxis potential.


Asunto(s)
Difosfonatos/farmacología , Fructosa-Bifosfato Aldolasa/antagonistas & inhibidores , Fructosa-Bifosfato Aldolasa/metabolismo , Animales , Dominio Catalítico , Difosfonatos/química , Diseño de Fármacos , Fructosa-Bifosfato Aldolasa/química , Glucólisis/efectos de los fármacos , Modelos Moleculares , Conejos
14.
FEBS Lett ; 592(10): 1667-1680, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29687459

RESUMEN

Type I phosphomannose isomerases (PMIs) are zinc-dependent monofunctional metalloenzymes catalysing the reversible isomerization of d-mannose 6-phosphate to d-fructose 6-phosphate. 5-Phospho-d-arabinonhydrazide (5PAHz), designed as an analogue of the enediolate high-energy intermediate, strongly inhibits PMI from Candida albicans (CaPMI). In this study, we report the 3D crystal structure of CaPMI complexed with 5PAHz at 1.85 Å resolution. The high-resolution structure suggests that Glu294 is the catalytic base that transfers a proton between the C1 and C2 carbon atoms of the substrate. Bidentate coordination of the inhibitor explains the stereochemistry of the isomerase activity, as well as the absence of both anomerase and C2-epimerase activities for Type I PMIs. A detailed mechanism of the reversible isomerization is proposed.


Asunto(s)
Candida albicans/enzimología , Hidrazinas/química , Manosa-6-Fosfato Isomerasa/química , Fosfatos de Azúcar/química , Dominio Catalítico , Cristalografía por Rayos X , Modelos Moleculares , Estructura Molecular , Estereoisomerismo , Especificidad por Sustrato
15.
Biosens Bioelectron ; 96: 178-185, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28500945

RESUMEN

Autocrine motility factor (AMF) is a tumor-secreted cytokine that stimulates tumor cell motility in vitro and metastasis in vivo. AMF could be detected in serum or urine of cancer patients with worse prognosis. Reported as a cancer biomarker, AMF secretion into body fluids might be closely related to metastases formation. In this study, a sensitive and specific carbohydrate-based electrochemical biosensor was designed for the detection and quantification of a protein model of AMF, namely phosphoglucose isomerase from rabbit muscle (RmPGI). Indeed, RmPGI displays high homology with AMF and has been shown to have AMF activity. The biosensor was constructed by covalent binding of the enzyme substrate d-fructose 6-phosphate (F6P). Immobilization was achieved on a gold surface electrode following a bottom-up approach through an aminated surface obtained by electrochemical patterning of ethylene diamine and terminal amine polyethylene glycol chain to prevent non-specific interactions. Carbohydrate-protein interactions were quantified in a range of 10 fM to 100nM. Complex formation was analyzed through monitoring of the redox couple Fe2+/Fe3+ by electrochemical impedance spectroscopy and square wave voltammetry. The F6P-biosensor demonstrates a detection limit of 6.6 fM and high selectivity when compared to other non-specific glycolytic proteins such as d-glucose-6-phosphate dehydrogenase. Detection of protein in spiked plasma was demonstrated and accuracy of 95% is obtained compared to result obtained in PBS (phosphate buffered saline). F6P-biosensor is a very promising proof of concept required for the design of a carbohydrate-based electrochemical biosensor using the enzyme substrate as bioreceptor. Such biosensor could be generalized to detect other protein biomarkers of interest.


Asunto(s)
Técnicas Biosensibles/métodos , Glucosa-6-Fosfato Isomerasa/sangre , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Técnicas Biosensibles/instrumentación , Espectroscopía Dieléctrica/instrumentación , Espectroscopía Dieléctrica/métodos , Diseño de Equipo , Fructosafosfatos/metabolismo , Glucosa-6-Fosfato Isomerasa/metabolismo , Oro/química , Humanos , Límite de Detección , Modelos Moleculares , Neoplasias/sangre , Neoplasias/metabolismo , Oxidación-Reducción , Conejos
16.
J Mol Biol ; 343(3): 649-57, 2004 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-15465052

RESUMEN

Pyrococcus furiosus phosphoglucose isomerase (PfPGI) is a metal-containing enzyme that catalyses the interconversion of glucose 6-phosphate (G6P) and fructose 6-phosphate (F6P). The recent structure of PfPGI has confirmed the hypothesis that the enzyme belongs to the cupin superfamily and identified the position of the active site. This fold is distinct from the alphabetaalpha sandwich fold commonly seen in phosphoglucose isomerases (PGIs) that are found in bacteria, eukaryotes and some archaea. Whilst the mechanism of the latter family is thought to proceed through a cis-enediol intermediate, analysis of the structure of PfPGI in the presence of inhibitors has led to the suggestion that the mechanism of this enzyme involves the metal-dependent direct transfer of a hydride between C1 and C2 atoms of the substrate. To gain further insight in the reaction mechanism of PfPGI, the structures of the free enzyme and the complexes with the inhibitor, 5-phospho-d-arabinonate (5PAA) in the presence and absence of metal have been determined. Comparison of these structures with those of equivalent complexes of the eukaryotic PGIs reveals similarities at the active site in the disposition of possible catalytic residues. These include the presence of a glutamic acid residue, Glu97 in PfPGI, which occupies the same position relative to the inhibitor as that of the glutamate that is thought to function as the catalytic base in the eukaryal-type PGIs. These similarities suggest that aspects of the catalytic mechanisms of these two structurally unrelated PGIs may be similar and based on an enediol intermediate.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Glucosa-6-Fosfato Isomerasa/química , Pentosafosfatos/química , Conformación Proteica , Pyrococcus furiosus/enzimología , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Dimerización , Fructosafosfatos/metabolismo , Glucosa-6-Fosfato/metabolismo , Manganeso/metabolismo , Modelos Moleculares , Estructura Molecular , Pentosafosfatos/metabolismo , Unión Proteica
17.
J Phys Chem B ; 115(25): 8304-16, 2011 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-21650197

RESUMEN

Using polarizable molecular mechanics, a recent study [de Courcy et al. J. Am. Chem. Soc., 2010, 132, 3312] has compared the relative energy balances of five competing inhibitors of the FAK kinase. It showed that the inclusion of structural water molecules was indispensable for an ordering consistent with the experimental one. This approach is now extended to compare the binding affinities of four active site ligands to the Type I Zn-metalloenzyme phosphomannose isomerase (PMI) from Candida albicans. The first three ones are the PMI substrate ß-D-mannopyranose 6-phosphate (ß-M6P) and two isomers, α-D-mannopyranose 6-phosphate (α-M6P) and ß-D-glucopyranose 6-phosphate (ß-G6P). They have a dianionic 6-phosphate substituent and differ by the relative configuration of the two carbon atoms C1 and C2 of the pyranose ring. The fourth ligand, namely 6-deoxy-6-dicarboxymethyl-ß-D-mannopyranose (ß-6DCM), is a substrate analogue that has the ß-M6P phosphate replaced by the nonhydrolyzable phosphate surrogate malonate. In the energy-minimized structures of all four complexes, one of the ligand hydroxyl groups binds Zn(II) through a water molecule, and the dianionic moiety binds simultaneously to Arg304 and Lys310 at the entrance of the cavity. Comparative energy-balances were performed in which solvation of the complexes and desolvation of PMI and of the ligands are computed using the Langlet-Claverie continuum reaction field procedure. They resulted into a more favorable balance in favor of ß-M6P than α-M6P and ß-G6P, consistent with the experimental results that show ß-M6P to act as a PMI substrate, while α-M6P and ß-G6P are inactive or at best weak inhibitors. However, these energy balances indicated the malonate ligand ß-6DCM to have a much lesser favorable relative complexation energy than the substrate ß-M6P, while it has an experimental 10-fold higher affinity than it on Type I PMI from Saccharomyces cerevisiae. The energy calculations were validated by comparison with parallel ab initio quantum chemistry on model binding sites extracted from the energy-minimized PMI-inhibitor complexes. We sought to improve the models upon including explicit water molecules solvating the dianionic moieties in their ionic bonds with the Arg304 and Lys310 side-chains. Energy-minimization resulted in the formation of three networks of structured waters. The first water of each network binds to one of the three accessible anionic oxygens. The networks extend to PMI residues (Asp17, Glu48, Asp300) remote from the ligand binding site. The final comparative energy balances also took into account ligand desolvation in a box of 64 waters. They now resulted into a large preference in favor of ß-6DCM over ß-M6P. The means to further augment the present model upon including entropy effects and sampling were discussed. Nevertheless a clear-cut conclusion emerging from this as well as our previous study on FAK kinase is that both polarization and charge-transfer contributions are critical elements of the energy balances.


Asunto(s)
Proteínas Fúngicas/química , Ligandos , Manosa-6-Fosfato Isomerasa/química , Manosafosfatos/química , Agua/química , Zinc/química , Secuencia de Aminoácidos , Candida albicans/enzimología , Isomerismo , Manosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/química , Alineación de Secuencia , Especificidad por Sustrato , Termodinámica
18.
Carbohydr Res ; 344(7): 869-80, 2009 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-19328460

RESUMEN

This study reports syntheses of d-allose 6-phosphate (All6P), D-allulose (or D-psicose) 6-phosphate (Allu6P), and seven D-ribose 5-phosphate isomerase (Rpi) inhibitors. The inhibitors were designed as analogues of the 6-carbon high-energy intermediate postulated for the All6P to Allu6P isomerization reaction (Allpi activity) catalyzed by type B Rpi from Escherichiacoli (EcRpiB). 5-Phospho-D-ribonate, easily obtained through oxidative cleavage of either All6P or Allu6P, led to the original synthon 5-dihydrogenophospho-D-ribono-1,4-lactone from which the other inhibitors could be synthesized through nucleophilic addition in one step. Kinetic evaluation on Allpi activity of EcRpiB shows that two of these compounds, 5-phospho-D-ribonohydroxamic acid and N-(5-phospho-D-ribonoyl)-methylamine, indeed behave as new efficient inhibitors of EcRpiB; further, 5-phospho-D-ribonohydroxamic acid was demonstrated to have competitive inhibition. Kinetic evaluation on Rpi activity of both EcRpiB and RpiB from Mycobacterium tuberculosis (MtRpiB) shows that several of the designed 6-carbon high-energy intermediate analogues are new competitive inhibitors of both RpiBs. One of them, 5-phospho-D-ribonate, not only appears as the strongest competitive inhibitor of a Rpi ever reported in the literature, with a K(i) value of 9 microM for MtRpiB, but also displays specific inhibition of MtRpiB versus EcRpiB.


Asunto(s)
Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Fosfatos de Azúcar/química , Isomerasas Aldosa-Cetosa/química , Dominio Catalítico , Inhibidores Enzimáticos/química , Ácidos Hidroxámicos/química , Cinética , Estructura Molecular , Mycobacterium tuberculosis/enzimología , Estructura Secundaria de Proteína
19.
J Mol Biol ; 382(3): 667-79, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18640127

RESUMEN

Interconversion of D-ribose-5-phosphate (R5P) and D-ribulose-5-phosphate is an important step in the pentose phosphate pathway. Two unrelated enzymes with R5P isomerase activity were first identified in Escherichia coli, RpiA and RpiB. In this organism, the essential 5-carbon sugars were thought to be processed by RpiA, while the primary role of RpiB was suggested to instead be interconversion of the rare 6-carbon sugars D-allose-6-phosphate (All6P) and D-allulose-6-phosphate. In Mycobacterium tuberculosis, where only an RpiB is found, the 5-carbon sugars are believed to be the enzyme's primary substrates. Here, we present kinetic studies examining the All6P isomerase activity of the RpiBs from these two organisms and show that only the E. coli enzyme can catalyze the reaction efficiently. All6P instead acts as an inhibitor of the M. tuberculosis enzyme in its action on R5P. X-ray studies of the M. tuberculosis enzyme co-crystallized with All6P and 5-deoxy-5-phospho-D-ribonohydroxamate (an inhibitor designed to mimic the 6-carbon sugar) and comparison with the E. coli enzyme's structure allowed us to identify differences in the active sites that explain the kinetic results. Two other structures, that of a mutant E. coli RpiB in which histidine 99 was changed to asparagine and that of wild-type M. tuberculosis enzyme, both co-crystallized with the substrate ribose-5-phosphate, shed additional light on the reaction mechanism of RpiBs generally.


Asunto(s)
Isomerasas Aldosa-Cetosa/metabolismo , Proteínas Bacterianas/metabolismo , Escherichia coli/enzimología , Glucosa/metabolismo , Mycobacterium tuberculosis/enzimología , Ribosamonofosfatos/metabolismo , Isomerasas Aldosa-Cetosa/química , Isomerasas Aldosa-Cetosa/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/metabolismo , Glucosa/química , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutación , Conformación Proteica , Ribosamonofosfatos/química
20.
J Mol Biol ; 379(2): 357-71, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18448118

RESUMEN

The archaeon Sulfolobus solfataricus converts d-arabinose to 2-oxoglutarate by an enzyme set consisting of two dehydrogenases and two dehydratases. The third step of the pathway is catalyzed by a novel 2-keto-3-deoxy-D-arabinonate dehydratase (KdaD). In this study, the crystal structure of the enzyme has been solved to 2.1 A resolution. The enzyme forms an oval-shaped ring of four subunits, each consisting of an N-terminal domain with a four-stranded beta-sheet flanked by two alpha-helices, and a C-terminal catalytic domain with a fumarylacetoacetate hydrolase (FAH) fold. Crystal structures of complexes of the enzyme with magnesium or calcium ions and either a substrate analog 2-oxobutyrate, or the aldehyde enzyme product 2,5-dioxopentanoate revealed that the divalent metal ion in the active site is coordinated octahedrally by three conserved carboxylate residues, a water molecule, and both the carboxylate and the oxo groups of the substrate molecule. An enzymatic mechanism for base-catalyzed dehydration is proposed on the basis of the binding mode of the substrate to the metal ion, which suggests that the enzyme enhances the acidity of the protons alpha to the carbonyl group, facilitating their abstraction by glutamate 114. A comprehensive structural comparison of members of the FAH superfamily is presented and their evolution is discussed, providing a basis for functional investigations of this largely unexplored protein superfamily.


Asunto(s)
Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Hidroliasas/química , Hidroliasas/metabolismo , Hidrolasas/química , Hidrolasas/metabolismo , Estructura Cuaternaria de Proteína , Secuencia de Aminoácidos , Animales , Proteínas Arqueales/genética , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Humanos , Hidroliasas/genética , Hidrolasas/genética , Magnesio/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína , Alineación de Secuencia , Especificidad por Sustrato , Sulfolobus solfataricus/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA