Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Chem Biol ; 28(11): 1554-1568.e8, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33915105

RESUMO

RNA-based sensors for intracellular metabolites are a promising solution to the emerging issue of metabolic heterogeneity. However, their development, i.e., the conversion of an aptamer into an in vivo-functional intracellular metabolite sensor, still harbors challenges. Here, we accomplished this for the glycolytic flux-signaling metabolite, fructose-1,6-bisphosphate (FBP). Starting from in vitro selection of an aptamer, we constructed device libraries with a hammerhead ribozyme as actuator. Using high-throughput screening in yeast with fluorescence-activated cell sorting (FACS), next-generation sequencing, and genetic-environmental perturbations to modulate the intracellular FBP levels, we identified a sensor that generates ratiometric fluorescent readout. An abrogated response in sensor mutants and occurrence of two sensor conformations-revealed by RNA structural probing-indicated in vivo riboswitching activity. Microscopy showed that the sensor can differentiate cells with different glycolytic fluxes within yeast populations, opening research avenues into metabolic heterogeneity. We demonstrate the possibility to generate RNA-based sensors for intracellular metabolites for which no natural metabolite-binding RNA element exits.


Assuntos
Técnicas Biossensoriais , Frutosedifosfatos/química , RNA/análise , Frutosedifosfatos/metabolismo , Glicólise , RNA/metabolismo , Saccharomyces cerevisiae/metabolismo
2.
Arch Biochem Biophys ; 695: 108633, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33075302

RESUMO

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.


Assuntos
Frutosedifosfatos/química , Fígado/enzimologia , Fosfoenolpiruvato/química , Piruvato Quinase/química , Regulação Alostérica , Frutosedifosfatos/metabolismo , Humanos , Fosfoenolpiruvato/metabolismo , Piruvato Quinase/metabolismo
3.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 6): 461-469, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-31204694

RESUMO

Human liver pyruvate kinase (hLPYK) converts phosphoenolpyruvate to pyruvate in the final step of glycolysis. hLPYK is allosterically activated by fructose-1,6-bisphosphate (Fru-1,6-BP). The allosteric site, as defined by previous structural studies, is located in domain C between the phosphate-binding loop (residues 444-449) and the allosteric loop (residues 527-533). In this study, the X-ray crystal structures of four hLPYK variants were solved to make structural correlations with existing functional data. The variants are D499N, W527H, Δ529/S531G (called GGG here) and S531E. The results revealed a conformational toggle between the open and closed positions of the allosteric loop. In the absence of Fru-1,6-BP the open position is stabilized, in part, by a cation-π bond between Trp527 and Arg538' (from an adjacent monomer). In the S531E variant glutamate binds in place of the 6'-phosphate of Fru-1,6-BP in the allosteric site, leading to partial allosteric activation. Finally, the structure of the D499N mutant does not provide structural evidence for the previously observed allosteric activation of the D499N variant.


Assuntos
Cátions/química , Frutosedifosfatos/metabolismo , Fígado/enzimologia , Mutação , Piruvato Quinase/química , Piruvato Quinase/metabolismo , Sítio Alostérico , Sítios de Ligação , Cristalografia por Raios X , Frutosedifosfatos/química , Humanos , Modelos Moleculares , Conformação Proteica , Subunidades Proteicas , Piruvato Quinase/genética
4.
Insect Biochem Mol Biol ; 104: 82-90, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30578824

RESUMO

Female Aedes aegypti mosquitoes are vectors of arboviruses that cause diseases of public health significance. The discovery of new metabolic targets is crucial for improving mosquito control strategies. We recently demonstrated that glucose oxidation supports ammonia detoxification in A. aegypti. Pyruvate kinase (PK, EC 2.7.1.40) catalyzes the last step of the glycolytic pathway. In most organisms, one or more allosteric effectors control PK activity. However, the kinetic properties and structure of PK in mosquitoes have not been previously reported. In this study, two alternatively spliced mRNA variants (AaPK1 and AaPK2) that code for PKs were identified in the A. aegypti genome. The AaPK1 mRNA variant, which encodes a 529 amino acid protein with an estimated molecular weight of ∼57 kDa, was cloned. The protein was expressed in Escherichia coli and purified. The AaPK1 kinetic properties were identified. The recombinant protein was also crystallized and its 3D structure determined. We found that alanine, glutamine, proline, serine and fructose-1-phosphate displayed a classic allosteric activation on AaPK1. Ribulose-5-phosphate acted as an allosteric inhibitor of AaPK1 but its inhibitory effect was reversed by alanine, glutamine, proline and serine. Additionally, the allosteric activation of AaPK1 by amino acids was weakened by fructose-1,6-bisphosphate, whereas the allosteric activation of AaPK1 by alanine and serine was diminished by glucose-6-phosphate. The AaPK1 structure shows the presence of fructose-1,6-bisphosphate in the allosteric site. Together, our results reveal that specific amino acids and phosphorylated sugars tightly regulate conformational dynamics and catalytic changes of AaPK1. The distinctive AaPK1 allosteric properties support a complex role for this enzyme within mosquito metabolism.


Assuntos
Aedes/enzimologia , Frutosedifosfatos/química , Glucose-6-Fosfato/química , Proteínas de Insetos/química , Piruvato Quinase/química , Aedes/genética , Regulação Alostérica/fisiologia , Processamento Alternativo/fisiologia , Animais , Feminino , Frutosedifosfatos/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Glucose-6-Fosfato/metabolismo , Proteínas de Insetos/biossíntese , Proteínas de Insetos/genética , Cinética , Domínios Proteicos , Piruvato Quinase/genética , Piruvato Quinase/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
J Chromatogr A ; 1522: 30-37, 2017 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-28958759

RESUMO

In this study, ZrO2 layer coated silica microspheres (ZrO2/SiO2) were successfully prepared by a facile one-step surfactant-free hydrothermal route under low pH condition. The synthesized ZrO2/SiO2 material was then modified with d-fructose 1, 6-bisphosphate (FDP) to improve the chromatographic separation property of the material. Fused-silica capillary columns were prepared with the modified material for evaluation. Phenolic, nucleobases and alkaloids compounds in hydrophilic interaction chromatographic (HILIC) mode showed symmetrical peaks. The FDP-ZrO2/SiO2 stationary phase showed better performance than ZrO2/SiO2 packing material and demonstrated great potential for application in HILIC mode.


Assuntos
Cromatografia Líquida/métodos , Frutosedifosfatos/química , Microesferas , Dióxido de Silício/química , Zircônio/química , Alcaloides/análise , Interações Hidrofóbicas e Hidrofílicas
6.
FEBS J ; 284(18): 2955-2980, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28715126

RESUMO

Changes in allosteric regulation of glycolytic enzymes have been linked to metabolic reprogramming involved in cancer. Remarkably, allosteric mechanisms control enzyme function at significantly shorter time-scales compared to the long-term effects of metabolic reprogramming on cell proliferation. It remains unclear if and how the speed and reversibility afforded by rapid allosteric control of metabolic enzymes is important for cell proliferation. Tools that allow specific, dynamic modulation of enzymatic activities in mammalian cells would help address this question. Towards this goal, we have used molecular dynamics simulations to guide the design of mPKM2 internal light/oxygen/voltage-sensitive domain 2 (LOV2) fusion at position D24 (PiL[D24]), an engineered pyruvate kinase M2 (PKM2) variant that harbours an insertion of the light-sensing LOV2 domain from Avena Sativa within a region implicated in allosteric regulation by fructose 1,6-bisphosphate (FBP). The LOV2 photoreaction is preserved in the PiL[D24] chimera and causes secondary structure changes that are associated with a 30% decrease in the Km of the enzyme for phosphoenolpyruvate resulting in increased pyruvate kinase activity after light exposure. Importantly, this change in activity is reversible upon light withdrawal. Expression of PiL[D24] in cells leads to light-induced increase in labelling of pyruvate from glucose. PiL[D24] therefore could provide a means to modulate cellular glucose metabolism in a remote manner and paves the way for studying the importance of rapid allosteric phenomena in the regulation of metabolism and enzyme control.


Assuntos
Apoproteínas/química , Proteínas de Transporte/química , Proteínas de Ligação a DNA/química , Frutosedifosfatos/química , Proteínas de Membrana/química , Proteínas de Plantas/química , Proteínas Recombinantes de Fusão/química , Hormônios Tireóideos/química , Regulação Alostérica , Sítio Alostérico , Motivos de Aminoácidos , Apoproteínas/genética , Apoproteínas/metabolismo , Avena/química , Avena/genética , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Frutosedifosfatos/metabolismo , Expressão Gênica , Humanos , Cinética , Luz , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Ligação Proteica , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Eletricidade Estática , Especificidade por Substrato , Termodinâmica , Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
7.
J Biol Chem ; 292(15): 6255-6268, 2017 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-28223362

RESUMO

ADP-glucose pyrophosphorylase (AGPase) controls bacterial glycogen and plant starch biosynthetic pathways, the most common carbon storage polysaccharides in nature. AGPase activity is allosterically regulated by a series of metabolites in the energetic flux within the cell. Very recently, we reported the first crystal structures of the paradigmatic AGPase from Escherichia coli (EcAGPase) in complex with its preferred physiological negative and positive allosteric regulators, adenosine 5'-monophosphate (AMP) and fructose 1,6-bisphosphate (FBP), respectively. However, understanding the molecular mechanism by which AMP and FBP allosterically modulates EcAGPase enzymatic activity still remains enigmatic. Here we found that single point mutations of key residues in the AMP-binding site decrease its inhibitory effect but also clearly abolish the overall AMP-mediated stabilization effect in wild-type EcAGPase. Single point mutations of key residues for FBP binding did not revert the AMP-mediated stabilization. Strikingly, an EcAGPase-R130A mutant displayed a dramatic increase in activity when compared with wild-type EcAGPase, and this increase correlated with a significant increment of glycogen content in vivo The crystal structure of EcAGPase-R130A revealed unprecedented conformational changes in structural elements involved in the allosteric signal transmission. Altogether, we propose a model in which the positive and negative energy reporters regulate AGPase catalytic activity via intra- and interprotomer cross-talk, with a "sensory motif" and two loops, RL1 and RL2, flanking the ATP-binding site playing a significant role. The information reported herein provides exciting possibilities for industrial/biotechnological applications.


Assuntos
Monofosfato de Adenosina/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Frutosedifosfatos/química , Glucose-1-Fosfato Adenililtransferase/química , Monofosfato de Adenosina/metabolismo , Regulação Alostérica , Cristalografia por Raios X , Estabilidade Enzimática , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Frutosedifosfatos/metabolismo , Glucose-1-Fosfato Adenililtransferase/genética , Glucose-1-Fosfato Adenililtransferase/metabolismo , Mutação Puntual
8.
Adv Exp Med Biol ; 925: 117-145, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27815924

RESUMO

Many bacterial L-lactate dehydrogenases (LDH) are allosteric enzymes, and usually activated by fructose 1,6-bisphosphate (FBP) and often also by substrate pyruvate. The active and inactive state structures demonstrate that Thermus caldophilus, Lactobacillus casei, and Bifidobacterium longum LDHs consistently undergo allosteric transition according to Monod-Wyman-Changeux model, where the active (R) and inactive (T) states of the enzymes coexist in an allosteric equilibrium (pre-existing equilibrium) independently of allosteric effectors. The three enzymes consistently take on open and closed conformations of the homotetramers for the T and R states, coupling the quaternary structural changes with the structural changes in binding sites for substrate and FBP though tertiary structural changes. Nevertheless, the three enzymes undergo markedly different structural changes from one another, indicating that there is a high variety in the allosteric machineries of bacterial LDHs. L. casei LDH undergoes the largest quaternary structural change in the three enzymes, and regulates its catalytic activity though a large linkage frame for allosteric motion. In contrast, T. caldophilus LDH exhibits the simplest allosteric motion in the three enzymes, involving a simple mobile structural core for the allosteric motion. TcLDH likely mediates its allosteric equilibrium mostly through electrostatic repulsion within the protein molecule, providing an insight for regulation machineries in bacterial allosteric LDHs.


Assuntos
Proteínas de Bactérias/química , Bifidobacterium longum/enzimologia , Frutosedifosfatos/química , L-Lactato Desidrogenase/química , Lacticaseibacillus casei/enzimologia , Ácido Pirúvico/química , Thermus/enzimologia , Regulação Alostérica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bifidobacterium longum/química , Bifidobacterium longum/genética , Sítios de Ligação , Frutosedifosfatos/metabolismo , Expressão Gênica , Cinética , L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/metabolismo , Lacticaseibacillus casei/química , Lacticaseibacillus casei/genética , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ácido Pirúvico/metabolismo , Especificidade da Espécie , Relação Estrutura-Atividade , Especificidade por Substrato , Thermus/química , Thermus/genética
9.
Structure ; 24(9): 1613-22, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27545622

RESUMO

ADP-glucose pyrophosphorylase (AGPase) catalyzes the rate-limiting step of bacterial glycogen and plant starch biosynthesis, the most common carbon storage polysaccharides in nature. A major challenge is to understand how AGPase activity is regulated by metabolites in the energetic flux within the cell. Here we report crystal structures of the homotetrameric AGPase from Escherichia coli in complex with its physiological positive and negative allosteric regulators, fructose-1,6-bisphosphate (FBP) and AMP, and sucrose in the active site. FBP and AMP bind to partially overlapping sites located in a deep cleft between glycosyltransferase A-like and left-handed ß helix domains of neighboring protomers, accounting for the fact that sensitivity to inhibition by AMP is modulated by the concentration of the activator FBP. We propose a model in which the energy reporters regulate EcAGPase catalytic activity by intra-protomer interactions and inter-protomer crosstalk, with a sensory motif and two regulatory loops playing a prominent role.


Assuntos
Proteínas de Escherichia coli/química , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Glucose-1-Fosfato Adenililtransferase/química , Glicogênio/biossíntese , Monofosfato de Adenosina/química , Monofosfato de Adenosina/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Frutosedifosfatos/química , Frutosedifosfatos/metabolismo , Glucose-1-Fosfato Adenililtransferase/genética , Glucose-1-Fosfato Adenililtransferase/metabolismo , Modelos Moleculares , Regiões Promotoras Genéticas , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Sacarose/química , Sacarose/metabolismo
10.
Biosens Bioelectron ; 83: 91-6, 2016 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-27107145

RESUMO

A new strategy to fabricate electrochemical biosensor is reported based on the linkage of enzyme substrate, thereby an electrochemical method to detect aldolase activity is established using pectin-thionine complex (PTC) as recognization element and signal probe. The linkage effect of fructose-1,6-bisphosphate (FBP), the substrate of aldolase, can be achieved via its strong binding to magnetic nanoparticles (MNPs)/aminophenylboronic acid (APBA) and the formation of phosphoramidate bond derived from its reaction with p-phenylenediamine (PDA) on the surface of electrode. Aldolase can reversibly catalyze the substrates into the products which have no binding capacity with MNPs/APBA, resulting in the exposure of the corresponding binding sites and its subsequent recognization on signal probe. Meanwhile, signal amplification can be accomplished by using the firstly prepared PTC which can bind with MNPs/APBA, and accuracy can be strengthened through magnetic separation. With good precision and accuracy, the established sensor may be extended to other proteins with reversible catalyzed ability.


Assuntos
Técnicas Eletroquímicas/métodos , Frutose-Bifosfato Aldolase/sangue , Frutose-Bifosfato Aldolase/metabolismo , Frutosedifosfatos/metabolismo , Pectinas/química , Fenotiazinas/química , Animais , Sítios de Ligação , Técnicas Biossensoriais/métodos , Ácidos Borônicos/química , Bovinos , Eletrodos , Ensaios Enzimáticos/métodos , Frutose-Bifosfato Aldolase/análise , Frutosedifosfatos/química , Nanopartículas de Magnetita/química , Coelhos , Especificidade por Substrato
11.
J Med Microbiol ; 65(1): 9-18, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26497196

RESUMO

Escherichia coli cra null mutants have been reported in the literature to be impaired in biofilm formation. To develop E. coli biofilm-inhibiting agents for prevention and control of adherent behaviour, analogues of a natural Cra ligand, fructose-1,6-bisphosphate, were identified based on two-dimensional similarity to the natural ligand. Of the analogues identified, those belonging to the bisphosphonate class of drug molecules were selected for study, as these are approved for clinical use in humans and their safety has been established. Computational and in vitro studies with purified Cra protein showed that risedronate sodium interacted with residues in the fructose-1,6-bisphosphate-binding site. Using a quantitative biofilm assay, risedronate sodium, at a concentration of 300-400 µM, was found to decrease E. coli and Salmonella pullorum biofilm formation by >60 %. Risedronate drastically reduced the adherence of E. coli cells to a rubber Foley urinary catheter, demonstrating its utility in preventing the formation of biofilm communities on medical implant surfaces. The use of risedronate, either alone or in combination with other agents, to prevent the formation of biofilms on surfaces is a novel finding that can easily be translated into practical applications.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Ácido Risedrônico/farmacologia , Proteínas de Bactérias/metabolismo , Clonagem Molecular , Frutosedifosfatos/química , Deleção de Genes , Proteínas Repressoras/metabolismo , Salmonella/efeitos dos fármacos , Cateterismo Urinário
12.
J Am Chem Soc ; 137(43): 13876-86, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26440863

RESUMO

Substrate recognition is one of the hallmarks of enzyme catalysis. Enzyme conformational changes have been linked to selectivity between substrates with little direct evidence. Aldolase, a glycolytic enzyme, must distinguish between two physiologically important substrates, fructose 1-phosphate and fructose 1,6-bisphosphate, and provides an excellent model system for the study of this question. Previous work has shown that isozyme specific residues (ISRs) distant from the active site are responsible for kinetic distinction between these substrates. Notably, most of the ISRs reside in a cluster of five surface α-helices, and the carboxyl-terminal region (CTR), and cooperative interactions among these helices have been demonstrated. To test the hypothesis that conformational changes are at the root of these changes, single surface-cysteine variants were created with the cysteine located on helices of the cluster and CTR. This allowed for site-specific labeling with an environmentally sensitive fluorophore, and subsequent monitoring of conformational changes by fluorescence emission spectrophotometry. These labeled variants revealed different spectra in the presence of saturating amounts of each substrate, which suggested the occurrence of different conformations. Emission spectra collected at various substrate concentrations showed a concentration dependence of the fluorescence spectra, consistent with binding events. Lastly, stopped-flow fluorescence spectrophotometry showed that the rate of these fluorescence changes was on the same time-scale as catalysis, thus suggesting a link between the different fluorescence changes and events during catalysis. On the basis of these results, we propose that different conformational changes may be a common mechanism for dictating substrate specificity in other enzymes with multiple substrates.


Assuntos
Frutose-Bifosfato Aldolase/química , Frutose-Bifosfato Aldolase/metabolismo , Frutosedifosfatos/química , Frutosedifosfatos/metabolismo , Frutosefosfatos/química , Frutosefosfatos/metabolismo , Cinética , Modelos Moleculares , Conformação Proteica , Espectrometria de Fluorescência , Especificidade por Substrato
13.
Biometals ; 28(4): 687-91, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25940829

RESUMO

Role of fructose 1,6-bisphosphate-mediated iron oxidation in the generation of reactive oxygen species was analyzed. Aconitase the most sensitive enzyme to oxidative stress was inactivated potently by fructose 1,6-bisphosphate in the presence of ferrous ion, and further by ADP and PEP to a lesser extent. The inactivation requires cyanide, suggesting that the superoxide radical is responsible for the inactivation. Addition of ascorbic acid and dithiothreitol prevented aconitase from the inactivation. Fructose 1,6-bisphosphate, ADP and PEP stimulated the oxidation of ferrous ion causing one-electron reduction of oxygen molecule. Superoxide radical formed with iron oxidation participates in the oxidative inactivation of aconitase and the citric acid cycle, resulting in the induction of the Crabtree effect, that is, high glucose-mediated inhibition of oxidative metabolism in mitochondria.


Assuntos
Aconitato Hidratase/metabolismo , Compostos Ferrosos/metabolismo , Frutosedifosfatos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Compostos Ferrosos/química , Frutosedifosfatos/química , Oxirredução , Saccharomyces cerevisiae/enzimologia
14.
Protein Cell ; 6(4): 275-287, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25645022

RESUMO

Pyruvate kinase isoform M2 (PKM2) converts phosphoenolpyruvate (PEP) to pyruvate and plays an important role in cancer metabolism. Here, we show that post-translational modifications and a patient-derived mutation regulate pyruvate kinase activity of PKM2 through modulating the conformation of the PKM2 tetramer. We determined crystal structures of human PKM2 mutants and proposed a "seesaw" model to illustrate conformational changes between an inactive T-state and an active R-state tetramers of PKM2. Biochemical and structural analyses demonstrate that PKM2(Y105E) (phosphorylation mimic of Y105) decreases pyruvate kinase activity by inhibiting FBP (fructose 1,6-bisphosphate)-induced R-state formation, and PKM2(K305Q) (acetylation mimic of K305) abolishes the activity by hindering tetramer formation. K422R, a patient-derived mutation of PKM2, favors a stable, inactive T-state tetramer because of strong intermolecular interactions. Our study reveals the mechanism for dynamic regulation of PKM2 by post-translational modifications and a patient-derived mutation and provides a structural basis for further investigation of other modifications and mutations of PKM2 yet to be discovered.


Assuntos
Proteínas de Transporte/química , Proteínas de Membrana/química , Neoplasias/enzimologia , Processamento de Proteína Pós-Traducional , Subunidades Proteicas/química , Hormônios Tireóideos/química , Acetilação , Regulação Alostérica , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Frutosedifosfatos/química , Frutosedifosfatos/metabolismo , Expressão Gênica , Humanos , Cinética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Mutação , Neoplasias/genética , Neoplasias/patologia , Fosforilação , Conformação Proteica , Multimerização Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Células Tumorais Cultivadas , Proteínas de Ligação a Hormônio da Tireoide
15.
Biochemistry ; 54(7): 1516-24, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25629396

RESUMO

In the study of allosteric proteins, understanding which effector-protein interactions contribute to allosteric activation is important both for designing allosteric drugs and for understanding allosteric mechanisms. The antihyperglycemic target, human liver pyruvate kinase (hL-PYK), binds its allosteric activator, fructose 1,6-bisphosphate (Fru-1,6-BP), such that the 1'-phosphate interacts with side chains of Arg501 and Trp494 and the 6'-phosphate interacts with Thr444, Thr446, Ser449 (i.e., the 444-449 loop), and Ser531. Additionally, backbone atoms from the 527-533 loop interact with a sugar ring hydroxyl and the two effector phosphate moieties. An effector analogue series indicates that only one phosphate on the sugar is required for activation. However, singly phosphorylated sugars, including Fru-1-P and Fru-6-P, bind with a Kix in the range of 0.07-1 mM. The second phosphate of Fru-1,6-BP causes tight effector binding, because this native effector has a Kix of 0.061 µM. Glucose 1,6-bisphosphate and ribulose 1,5-bisphosphate bind in the 0.07-1 mM range. The contrast with a higher Fru-1,6-BP binding indicates specificity for the fructose sugar conformation. Site-directed random mutagenesis at each residue that contacts bound Fru-1,6-BP showed that a negative charge introduced at position 531 mimics allosteric activation, even in the absence of Fru-1,6-BP. Collectively, analogue and mutagenesis studies are consistent with the 527-533 loop playing a key role in allosteric function. Deletion mutations that shortened the 527-533 loop were expected to prevent formation of hydrogen bonds between backbone atoms on the loop and Fru-1,6-BP. Indeed, Fru-1,6-BP did not activate these loop-shortened mutant proteins. Previous structural comparisons of M1-PYK and M2-PYK indicate that the 527-533 loop makes interactions across a subunit interface when an activator is not present. Mutating the hL-PYK subunit interface interactions among Trp527, Arg528, and Asp499 mimics allosteric activation. Considered with published structures, these results are consistent with (1) the two phosphates of Fru-1,6-BP docking to Arg501/Trp494 and the 444-449 loop, respectively, and (2) the formation of hydrogen bonds among Fru-1,6-BP and backbone atoms of the 527-533 loop pulling this loop away from the subunit interface, which results in breaking of the Trp527-Arg528-Asp499 interactions to elicit an allosteric response.


Assuntos
Frutosedifosfatos/metabolismo , Fígado/enzimologia , Piruvato Quinase/química , Piruvato Quinase/metabolismo , Regulação Alostérica , Sítios de Ligação , Frutosedifosfatos/química , Humanos , Modelos Moleculares , Mutagênese , Mutação , Piruvato Quinase/genética
16.
J Biol Chem ; 289(45): 31550-64, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25258319

RESUMO

For Thermus caldophilus L-lactate dehydrogenase (TcLDH), fructose 1,6-bisphosphate (FBP) reduced the pyruvate S(0.5) value 10(3)-fold and increased the V(max) value 4-fold at 30 °C and pH 7.0, indicating that TcLDH has a much more T state-sided allosteric equilibrium than Thermus thermophilus L-lactate dehydrogenase, which has only two amino acid replacements, A154G and H179Y. The inactive (T) and active (R) state structures of TcLDH were determined at 1.8 and 2.0 Å resolution, respectively. The structures indicated that two mobile regions, MR1 (positions 172-185) and MR2 (positions 211-221), form a compact core for allosteric motion, and His(179) of MR1 forms constitutive hydrogen bonds with MR2. The Q4(R) mutation, which comprises the L67E, H68D, E178K, and A235R replacements, increased V(max) 4-fold but reduced pyruvate S(0.5) only 5-fold in the reaction without FBP. In contrast, the P2 mutation, comprising the R173Q and R216L replacements, did not markedly increase V(max), but 10(2)-reduced pyruvate S(0.5), and additively increased the FBP-independent activity of the Q4(R) enzyme. The two types of mutation consistently increased the thermal stability of the enzyme. The MR1-MR2 area is a positively charged cluster, and its center approaches another positively charged cluster (N domain cluster) across the Q-axis subunit interface by 5 Å, when the enzyme undergoes the T to R transition. Structural and kinetic analyses thus revealed the simple and unique allosteric machinery of TcLDH, where the MR1-MR2 area pivotally moves during the allosteric motion and mediates the allosteric equilibrium through electrostatic repulsion within the protein molecule.


Assuntos
Proteínas de Bactérias/química , L-Lactato Desidrogenase/química , Thermus/enzimologia , Regulação Alostérica , Sítio Alostérico , Sequência de Aminoácidos , Catálise , Domínio Catalítico , Frutosedifosfatos/química , Concentração de Íons de Hidrogênio , Ácido Láctico/química , Dados de Sequência Molecular , Movimento (Física) , Mutação , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Ácido Pirúvico/química , Homologia de Sequência de Aminoácidos , Eletricidade Estática
17.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 9): 1186-92, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25195889

RESUMO

The apicomplexan parasite Toxoplasma gondii must invade host cells to continue its lifecycle. It invades different cell types using an actomyosin motor that is connected to extracellular adhesins via the bridging protein fructose-1,6-bisphosphate aldolase. During invasion, aldolase serves in the role of a structural bridging protein, as opposed to its normal enzymatic role in the glycolysis pathway. Crystal structures of the homologous Plasmodium falciparum fructose-1,6-bisphosphate aldolase have been described previously. Here, T. gondii fructose-1,6-bisphosphate aldolase has been crystallized in space group P22121, with the biologically relevant tetramer in the asymmetric unit, and the structure has been determined via molecular replacement to a resolution of 2.0 Å. An analysis of the quality of the model and of the differences between the four chains in the asymmetric unit and a comparison between the T. gondii and P. falciparum aldolase structures is presented.


Assuntos
Frutose-Bifosfato Aldolase/química , Frutosedifosfatos/química , Toxoplasma/enzimologia , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Dados de Sequência Molecular , Conformação Proteica
18.
J Mol Model ; 20(9): 2447, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25208557

RESUMO

Tyrosine phosphorylation (p-Y105) of pyruvate kinase (PK) M2, in recent years, has been suggested to facilitate Warburg effect and tumor cell growth. However, a comparison of the structural dynamics of the un-phosphorylated, the active, and the phosphorylated-at-Y105, the inactive-states, is not clear. We studied molecular dynamics of the two states to unravel these features, where phosphorylated PKM2 showed a rapid global conformation change in the initial stages of the simulation. The overall simulation identified that the phosphorylation event results in more buried and less flexible PKM2 conformation, as compared to the un-phosphorylated form, resulting in an open and closed conformation of the active site in un-phosphorylated and phosphorylated forms, respectively, due to the movement of B domain. This conformational shift in Y105-phosphorylated-PKM2 (p-Y105-PKM2) with closed active site, responsible for inhibition of PKM2 activity, was an outcome of the bending residues (117-118, 218-219, 296-297, and 301-308) within the loop connecting A and B domains and the presence of helix-loop-helix motif in A domain. The un-phosphorylated PKM2 formed a helix bend (H4) due to less fluctuation of the residue S-100; where the other end of the helix (H4) was connected to the substrate binding pocket. Further, simulation analysis showed that phosphorylation did not affect the FBP binding predominantly. We propose that p-Y105 inhibits the activity of PKM2 without influencing FBP binding directly and not allowing the open binding conformation by influencing G128, S100, G506 and gamma turn, G126 and S127 residues. Phosphorylated PKM2 was also identified to gain the transcriptional factor function which was not the case with un-phosphorylated form. These structurally important residues in PKM2 could have a bearing on cancer metabolism, since PKM2 has been implicated in the promotion of cancer in the recent past.


Assuntos
Simulação de Dinâmica Molecular , Piruvato Quinase/química , Sítios de Ligação , Domínio Catalítico , Ativação Enzimática , Frutosedifosfatos/química , Frutosedifosfatos/metabolismo , Cinética , Fosforilação , Ligação Proteica , Conformação Proteica , Piruvato Quinase/metabolismo , Relação Estrutura-Atividade , Tirosina
19.
PLoS One ; 9(8): e103888, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25102309

RESUMO

Fructose-1,6-bisphosphate activates ADP-glucose pyrophosphorylase and the synthesis of glycogen in Escherichia coli. Here, we show that although pyruvate is a weak activator by itself, it synergically enhances the fructose-1,6-bisphosphate activation. They increase the enzyme affinity for each other, and the combination increases Vmax, substrate apparent affinity, and decreases AMP inhibition. Our results indicate that there are two distinct interacting allosteric sites for activation. Hence, pyruvate modulates E. coli glycogen metabolism by orchestrating a functional network of allosteric regulators. We postulate that this novel dual activator mechanism increases the evolvability of ADP-glucose pyrophosphorylase and its related metabolic control.


Assuntos
Escherichia coli/enzimologia , Glucose-1-Fosfato Adenililtransferase/metabolismo , Piruvatos/metabolismo , Sítio Alostérico , Ativação Enzimática , Frutosedifosfatos/química , Frutosedifosfatos/metabolismo , Glicogênio/biossíntese , Cinética , Piruvatos/química , Especificidade por Substrato
20.
Elife ; 32014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25009227

RESUMO

Aerobic glycolysis or the Warburg Effect (WE) is characterized by the increased metabolism of glucose to lactate. It remains unknown what quantitative changes to the activity of metabolism are necessary and sufficient for this phenotype. We developed a computational model of glycolysis and an integrated analysis using metabolic control analysis (MCA), metabolomics data, and statistical simulations. We identified and confirmed a novel mode of regulation specific to aerobic glycolysis where flux through GAPDH, the enzyme separating lower and upper glycolysis, is the rate-limiting step in the pathway and the levels of fructose (1,6) bisphosphate (FBP), are predictive of the rate and control points in glycolysis. Strikingly, negative flux control was found and confirmed for several steps thought to be rate-limiting in glycolysis. Together, these findings enumerate the biochemical determinants of the WE and suggest strategies for identifying the contexts in which agents that target glycolysis might be most effective.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Biologia Computacional/métodos , Frutosedifosfatos/química , Glucose/química , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glicólise , Células HCT116 , Humanos , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/química , Espectrometria de Massas , Metabolômica/métodos , Modelos Químicos , Método de Monte Carlo , Fenótipo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA