Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
J Biol Chem ; 296: 100628, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33812994

RESUMEN

Catalysis of human phosphoglycerate mutase is dependent on a 2,3-bisphosphoglycerate cofactor (dPGM), whereas the nonhomologous isozyme in many parasitic species is cofactor independent (iPGM). This mechanistic and phylogenetic diversity offers an opportunity for selective pharmacologic targeting of glycolysis in disease-causing organisms. We previously discovered ipglycermide, a potent inhibitor of iPGM, from a large combinatorial cyclic peptide library. To fully delineate the ipglycermide pharmacophore, herein we construct a detailed structure-activity relationship using 280 substituted ipglycermide analogs. Binding affinities of these analogs to immobilized Caenorhabditis elegans iPGM, measured as fold enrichment relative to the index residue by deep sequencing of an mRNA display library, illuminated the significance of each amino acid to the pharmacophore. Using cocrystal structures and binding kinetics, we show that the high affinity of ipglycermide for iPGM orthologs, from Brugia malayi, Onchocerca volvulus, Dirofilaria immitis, and Escherichia coli, is achieved by a codependence between (1) the off-rate mediated by the macrocycle Cys14 thiolate coordination to an active-site Zn2+ in the iPGM phosphatase domain and (2) shape complementarity surrounding the macrocyclic core at the phosphotransferase-phosphatase domain interface. Our results show that the high-affinity binding of ipglycermide to iPGMs freezes these structurally dynamic enzymes into an inactive, stable complex.


Asunto(s)
Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Animales , Dominio Catalítico , Humanos , Modelos Moleculares , Filogenia , Conformación Proteica , Relación Estructura-Actividad
2.
Proteins ; 89(11): 1541-1556, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34245187

RESUMEN

The expansion of three-dimensional protein structures and enhanced computing power have significantly facilitated our understanding of protein sequence/structure/function relationships. A challenge in structural genomics is to predict the function of uncharacterized proteins. Protein function deconvolution based on global sequence or structural homology is impracticable when a protein relates to no other proteins with known function, and in such cases, functional relationships can be established by detecting their local ligand binding site similarity. Here, we introduce a sequence order-independent comparison algorithm, PocketShape, for structural proteome-wide exploration of protein functional site by fully considering the geometry of the backbones, orientation of the sidechains, and physiochemical properties of the pocket-lining residues. PocketShape is efficient in distinguishing similar from dissimilar ligand binding site pairs by retrieving 99.3% of the similar pairs while rejecting 100% of the dissimilar pairs on a dataset containing 1538 binding site pairs. This method successfully classifies 83 enzyme structures with diverse functions into 12 clusters, which is highly in accordance with the actual structural classification of proteins classification. PocketShape also achieves superior performances than other methods in protein profiling based on experimental data. Potential new applications for representative SARS-CoV-2 drugs Remdesivir and 11a are predicted. The high accuracy and time-efficient characteristics of PocketShape will undoubtedly make it a promising complementary tool for proteome-wide protein function inference and drug repurposing study.


Asunto(s)
Algoritmos , Antivirales/farmacología , Reposicionamiento de Medicamentos/métodos , Proteínas/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/química , Alanina/metabolismo , Alanina/farmacología , Antivirales/química , Sitios de Unión , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/metabolismo , Bases de Datos de Proteínas , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/metabolismo , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Proteínas/química , Proteínas/clasificación , Curva ROC , SARS-CoV-2/efectos de los fármacos
3.
Biochemistry ; 56(50): 6555-6564, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29166011

RESUMEN

Previously, we demonstrated that binding of a ligand to Escherichia coli cofactor-dependent phosphoglycerate mutase (dPGM), a homodimeric protein, is energetically coupled with dimerization. The equilibrium unfolding of dPGM occurs with a stable, monomeric intermediate. Binding of several nonsubstrate metabolites stabilizes the dimeric native form over the monomeric intermediate, reducing the population of the intermediate. Both the active site and the dimer interface appear to be unfolded in the intermediate. We hypothesized that a loop containing residues 118-152 was responsible for the energetic coupling between the dimer interface and the distal active site and was unfolded in the intermediate. Here, we investigated the structure of the dPGM intermediate by probing side-chain interactions and solvent accessibility of the peptide backbone. By comparing the effect of a mutation on the global stability and the stability of the intermediate, we determine an equilibrium φ value (φeq value), which provides information about whether side-chain interactions are retained or lost in the intermediate. Hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS) was used to investigate differences in the solvent accessibility of the peptide backbone in the intermediate and native forms of dPGM. The results of φeq value analysis and HDX-MS reveal the least stable folding unit of dPGM, which is unfolded in the intermediate and links the active site to the dimer interface. The structure of the intermediate reveals how the cooperative network of residues in dPGM gives rise to the observed energetic coupling between dimerization and ligand binding.


Asunto(s)
Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Secuencia de Aminoácidos , Deuterio , Dimerización , Escherichia coli/química , Proteínas de Escherichia coli/química , Hidrógeno , Ligandos , Espectrometría de Masas/métodos , Modelos Moleculares , Fosfoglicerato Mutasa/genética , Unión Proteica/fisiología , Conformación Proteica , Desnaturalización Proteica , Pliegue de Proteína
4.
Acta Pharmacol Sin ; 38(12): 1673-1682, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28748916

RESUMEN

Phosphoglycerate mutase 1 (PGAM1), an important enzyme in glycolysis, is overexpressed in a number of human cancers, thus has been proposed as a promising metabolic target for cancer treatments. The C-terminal portion of the available crystal structures of PGAM1 and its homologous proteins is partially disordered, as evidenced by weak electron density. In this study, we identified the conformational behavior of the C-terminal region of PGAM1 as well as its role during the catalytic cycle. Using the PONDR-FIT server, we demonstrated that the C-terminal region was intrinsically disordered. We applied the Monte Carlo (MC) method to explore the conformational space of the C-terminus and conducted a series of explicit-solvent molecular dynamics (MD) simulations, and revealed that the C-terminal region is inherently dynamic; large-scale conformational changes in the C-terminal segment led to the structural transition of PGAM1 from the closed state to the open state. Furthermore, the C-terminal segment influenced 2,3-bisphosphoglycerate (2,3-BPG) binding. The proposed swing model illustrated a critical role of the C-terminus in the catalytic cycle through the conformational changes. In conclusion, the C-terminal region induces large movements of PGAM1 from the closed state to the open state and influences cofactor binding during the catalytic cycle. This report describes the dynamic features of the C-terminal region in detail and should aid in design of novel and efficient inhibitors of PGAM1. A swing mechanism of the C-terminal region is proposed, to facilitate further studies of the catalytic mechanism and the physiological functions of its homologues.


Asunto(s)
Simulación de Dinámica Molecular , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Biocatálisis , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Método de Montecarlo , Fosfoglicerato Mutasa/antagonistas & inhibidores , Análisis de Componente Principal , Conformación Proteica , Electricidad Estática
5.
Biochemistry ; 55(12): 1711-23, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26919584

RESUMEN

Energetic coupling of two molecular events in a protein molecule is ubiquitous in biochemical reactions mediated by proteins, such as catalysis and signal transduction. Here, we investigate energetic coupling between ligand binding and folding of a dimer using a model system that shows three-state equilibrium unfolding of an exceptional quality. The homodimeric Escherichia coli cofactor-dependent phosphoglycerate mutase (dPGM) was found to be stabilized by ATP in a proteome-wide screen, although dPGM does not require or utilize ATP for enzymatic function. We investigated the effect of ATP on the thermodynamic stability of dPGM using equilibrium unfolding. We found that, in the absence of ATP, dPGM populates a partially unfolded, monomeric intermediate during equilibrium unfolding. However, addition of 1.0 mM ATP drastically reduces the population of the intermediate by selectively stabilizing the native dimer. Using a computational ligand docking method, we predicted ATP binds to the active site of the enzyme using the triphosphate group. By performing equilibrium unfolding and isothermal titration calorimetry with active-site variants of dPGM, we confirmed that active-site residues are involved in ATP binding. Our findings show that ATP promotes dimerization of the protein by binding to the active site, which is distal from the dimer interface. This cooperativity suggests an energetic coupling between the active site and the dimer interface. We also propose a structural link to explain how ligand binding to the active site is energetically coupled with dimerization.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Multimerización de Proteína/fisiología , Cristalografía por Rayos X , Ligandos , Unión Proteica/fisiología , Estructura Secundaria de Proteína
6.
Biochem Biophys Res Commun ; 450(2): 936-41, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24984149

RESUMEN

Three structurally distinct forms of phosphoglycerate mutase from the trypanosomatid parasite Leishmania mexicana were isolated by standard procedures of bacterial expression and purification. Analytical size-exclusion chromatography coupled to a multi-angle scattering detector detected two monomeric forms of differing hydrodynamic radii, as well as a dimeric form. Structural comparisons of holoenzyme and apoenzyme trypanosomatid cofactor-independent phosphoglycerate mutase (iPGAM) X-ray crystal structures show a large conformational change between the open (apoenzyme) and closed (holoenzyme) forms accounting for the different monomer hydrodynamic radii. Until now iPGAM from trypanosomatids was considered to be only monomeric, but results presented here show the appearance of a dimeric form. Taken together, these observations are important for the choice of screening strategies to identify inhibitors of iPGAM for parasite chemotherapy and highlight the need to select the most biologically or functionally relevant form of the purified enzyme.


Asunto(s)
Leishmania mexicana/enzimología , Fosfoglicerato Mutasa/química , Apoenzimas/química , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Cristalografía por Rayos X , Holoenzimas/química , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Especificidad por Sustrato
7.
J Cell Physiol ; 227(6): 2613-21, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22367961

RESUMEN

Cancer cells prefer anaerobic ATP synthesis, regardless of the availability of oxygen. It has been hypothesized that in these cells, glycolytic enzymes associate into a large complex, which results in an increased efficiency of glycolytic flux. However, there is no convincing in vivo evidence supporting this hypothesis. Here, we show that all the enzymes of triose phosphate metabolism, from aldolase to pyruvate kinase consecutively, form a macromolecular complex in vivo and that disruption of such complex significantly inhibits lactate release and ATP synthesis in the glycolytic pathway. Composition of the complex and the effectiveness of the glycolytic flux depends on lactate and glucose concentration. High concentrations of exogenous lactate reduces association of the C-terminal region phosphoglycerate mutase (PGAM) with the complex which results in its disruption and inhibition of ATP synthesis. Additionally, high lactate affects nuclear localization of PGAM and ceases cell proliferation. Our findings might provide new prospects for cancer treatment using low-molecular weight competitors to destabilize the glycolytic complex and reduce proliferative potential of cancer cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Células Escamosas/enzimología , Metabolismo Energético , Glucólisis , Neoplasias Pulmonares/enzimología , Fosfoglicerato Mutasa/metabolismo , Transporte Activo de Núcleo Celular , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Metabolismo Energético/efectos de los fármacos , Fructosa-Bifosfato Aldolasa/metabolismo , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Ácido Láctico/metabolismo , Neoplasias Pulmonares/patología , Ratones , Complejos Multienzimáticos , Péptidos/farmacología , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/farmacología , Estructura Terciaria de Proteína , Piruvato Quinasa/metabolismo
8.
Sci Data ; 9(1): 614, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36220829

RESUMEN

We have previously shown that in Arabidopsis the three enzymes of lower glycolysis namely phosphoglycerate mutase (PGAM), enolase and pyruvate kinase form a complex which plays an important role in tethering the mitochondria to the chloroplast. Given that the metabolism of these mutants, the complemented of pgam mutant and overexpression lines of PGAM were unclear, here, we present gas chromatography mass spectrometry-based metabolomics data of them alongside their plant growth phenotypes. Compared with wild type, both sugar and amino acid concentration are significantly altered in phosphoglycerate mutase, enolase and pyruvate kinase. Conversely, overexpression of PGAM could decrease the content of 3PGA, sugar and several amino acids and increase the content of alanine and pyruvate. In addition, the pgam mutant could not be fully complemented by either a nuclear target pgam, a side-directed-mutate of pgam or a the E.coli PGAM in term of plant phenotype or metabolite profiles, suggesting the low glycolysis complete formation is required to support normal metabolism and growth.


Asunto(s)
Arabidopsis , Alanina/metabolismo , Aminoácidos , Arabidopsis/genética , Arabidopsis/metabolismo , Glucólisis , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/genética , Fosfoglicerato Mutasa/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Piruvato Quinasa/metabolismo , Piruvatos , Azúcares
9.
J Exp Bot ; 62(14): 5179-89, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21813794

RESUMEN

Stomatal movements require massive changes in guard cell osmotic content, and both stomatal opening and stomatal closure have been shown to be energy-requiring processes. A possible role for glycolysis in contributing to the energetic, reducing requirements, or signalling processes regulating stomatal movements has not been investigated previously. Glycolysis, oxidization of glucose to pyruvate, is a central metabolic pathway and yields a net gain of 2 ATP and 2 NADH. 2,3-biphosphoglycerate-independent phosphoglycerate mutase (iPGAM) is a key enzymatic activity in glycolysis and catalyses the reversible interconversion of 3-phosphoglycerate to 2-phosphoglycerate. To investigate functions of iPGAMs and glycolysis in stomatal function and plant growth, Arabidopsis insertional mutants in At1g09780 and At3g08590, both of which have been annotated as iPGAMs on the basis of sequence homology, were identified and characterized. While single mutants were indistinguishable from the wild type in all plant phenotypes assayed, double mutants had no detectable iPGAM activity and showed defects in blue light-, abscisic acid-, and low CO(2)-regulated stomatal movements. Vegetative plant growth was severely impaired in the double mutants and pollen was not produced. The data demonstrate that iPGAMs and glycolytic activity are critical for guard cell function and fertility in Arabidopsis.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Fosfoglicerato Mutasa/metabolismo , Estomas de Plantas/fisiología , Polen/crecimiento & desarrollo , Secuencia de Aminoácidos , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Arabidopsis/fisiología , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Ácidos Glicéricos/metabolismo , Glucólisis , Datos de Secuencia Molecular , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/genética , Estomas de Plantas/enzimología , Estomas de Plantas/genética , Estomas de Plantas/crecimiento & desarrollo , Polen/enzimología , Polen/genética , Polen/metabolismo , Alineación de Secuencia
10.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 9): 1044-50, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21904048

RESUMEN

Burkholderia pseudomallei is a soil-dwelling bacterium endemic to Southeast Asia and Northern Australia. Burkholderia is responsible for melioidosis, a serious infection of the skin. The enzyme 2,3-bisphosphoglycerate-dependent phosphoglycerate mutase (PGAM) catalyzes the interconversion of 3-phosphoglycerate and 2-phosphoglycerate, a key step in the glycolytic pathway. As such it is an extensively studied enzyme and X-ray crystal structures of PGAM enzymes from multiple species have been elucidated. Vanadate is a phosphate mimic that is a powerful tool for studying enzymatic mechanisms in phosphoryl-transfer enzymes such as phosphoglycerate mutase. However, to date no X-ray crystal structures of phosphoglycerate mutase have been solved with vanadate acting as a substrate mimic. Here, two vanadate complexes together with an ensemble of substrate and fragment-bound structures that provide a comprehensive picture of the function of the Burkholderia enzyme are reported.


Asunto(s)
Burkholderia pseudomallei/enzimología , Fosfoglicerato Mutasa/química , Cristalografía por Rayos X , Modelos Moleculares , Fosfoglicerato Mutasa/metabolismo , Estructura Terciaria de Proteína , Especificidad por Sustrato
11.
Eur J Med Genet ; 64(9): 104283, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34237446

RESUMEN

BACKGROUND: Phosphoglycerate mutase (PGAM) deficiency is associated with a rare glycogen storage disease (glycogenosis type X) in humans caused by pathogenic variants in the PGAM2 gene. Several genes causing autosomal forms of glycogen storage disease (GSD) have been identified, involved in various forms of neuromuscular anomalies. METHODS: Targeted whole exome sequencing (WES) was performed on the DNA of single affected individual (IV-1) followed by Sanger sequencing confirmation of the identified variant in all available members of the family. RESULTS: In the present study, the affected individual, presenting mild features of glycogen storage disease type X. Targeted exome sequencing revealed a biallelic frameshift variant (c.687dupC; p. Met230Hisfs*6) in the PGAM2 gene located on chromosome 7p13. CONCLUSION: In short, we reported a novel homozygous frameshift variant as a cause of glycogen storage disease type X from Pakistani population. The work presented here proves significance of targeted WES in accurate diagnosis of known complex genetic disorders.


Asunto(s)
Enfermedades Renales/genética , Enfermedades Musculares/genética , Fosfoglicerato Mutasa/deficiencia , Fosfoglicerato Mutasa/genética , Adolescente , Mutación del Sistema de Lectura , Homocigoto , Humanos , Enfermedades Renales/patología , Masculino , Enfermedades Musculares/patología , Fosfoglicerato Mutasa/química
12.
Int J Biol Macromol ; 178: 1-10, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33631257

RESUMEN

Entamoeba histolytica phosphoserine phosphatase (EhPSP), a regulatory enzyme in the serine biosynthetic pathway, is also a structural homolog of cofactor-dependent phosphoglycerate mutase (dPGM). However, despite sharing many of its catalytic residues with dPGM, EhPSP displays no significant mutase activity. In the current work, we determined a crystal structure of EhPSP in complex with 3-PGA to 2.5 Å resolution and observed striking differences between the orientation of 3-PGA bound to EhPSP and that to its other homologous structures. We also performed computational modeling and simulations of the intermediate 2,3-bisphosphoglyceric acid into the active site of EhPSP to better understand its mechanistic details. Based on these results and those of a similar study with the dPGMs from E. coli and B. pseudomallei, the affinity of EhPSP for 2,3-BPG was concluded to be lower than those of the other proteins. Moreover, a different set of 2,3-BPG interacting residues was observed in EhPSP compared to dPGMs, with all of the crucial interacting residues of dPGMs either missing or substituted with weakly interacting residues. This study has expanded our understanding, at the structural level, of the inability of EhPSP to catalyze the mutase reaction and has strengthened earlier conclusions indicating it to be a true phosphatase.


Asunto(s)
Entamoeba histolytica/enzimología , Ácidos Glicéricos/química , Fosfoglicerato Mutasa/química , Monoéster Fosfórico Hidrolasas/química , Proteínas Protozoarias/química , Dominio Catalítico , Modelos Moleculares , Conformación Proteica , Alineación de Secuencia
13.
PLoS One ; 16(3): e0241738, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33760815

RESUMEN

Naegleria fowleri is a pathogenic, thermophilic, free-living amoeba which causes primary amebic meningoencephalitis (PAM). Penetrating the olfactory mucosa, the brain-eating amoeba travels along the olfactory nerves, burrowing through the cribriform plate to its destination: the brain's frontal lobes. The amoeba thrives in warm, freshwater environments, with peak infection rates in the summer months and has a mortality rate of approximately 97%. A major contributor to the pathogen's high mortality is the lack of sensitivity of N. fowleri to current drug therapies, even in the face of combination-drug therapy. To enable rational drug discovery and design efforts we have pursued protein production and crystallography-based structure determination efforts for likely drug targets from N. fowleri. The genes were selected if they had homology to drug targets listed in Drug Bank or were nominated by primary investigators engaged in N. fowleri research. In 2017, 178 N. fowleri protein targets were queued to the Seattle Structural Genomics Center of Infectious Disease (SSGCID) pipeline, and to date 89 soluble recombinant proteins and 19 unique target structures have been produced. Many of the new protein structures are potential drug targets and contain structural differences compared to their human homologs, which could allow for the development of pathogen-specific inhibitors. Five of the structures were analyzed in more detail, and four of five show promise that selective inhibitors of the active site could be found. The 19 solved crystal structures build a foundation for future work in combating this devastating disease by encouraging further investigation to stimulate drug discovery for this neglected pathogen.


Asunto(s)
Descubrimiento de Drogas , Naegleria fowleri/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Adenosilhomocisteinasa/antagonistas & inhibidores , Adenosilhomocisteinasa/química , Adenosilhomocisteinasa/metabolismo , Sitios de Unión , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Simulación de Dinámica Molecular , Naegleria fowleri/genética , Fosfoglicerato Mutasa/antagonistas & inhibidores , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Estructura Cuaternaria de Proteína , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteoma , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo
14.
Acta Biochim Pol ; 68(4): 515-525, 2021 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-34773933

RESUMEN

Although many atypical proteinaceous cell wall components that belong to a group of multitasking, "moonlighting" proteins, have been repeatedly identified in numerous pathogenic microorganisms, their novel extracellular functions and secretion mechanisms remain largely unrecognized. In Candida albicans, one of the most common fungal pathogens in humans, phosphoglycerate mutase (Gpm1) - a cytoplasmic enzyme involved in the glycolysis pathway - has been shown to occur on the cell surface and has been identified as a potentially important virulence factor. In this study, we demonstrated tight binding of C. albicans Gpm1 to the candidal cell surface, thus suggesting that the readsorption of soluble Gpm1 from the external environment could be a likely mechanism leading to the presence of this moonlighting protein on the pathogen surface. Several putative Gpm1-binding receptors on the yeast surface were identified. The affinities of Gpm1 to human vitronectin (VTR) and fibronectin (FN) were characterized with surface plasmon resonance measurements, and the dissociation constants of the complexes formed were determined to be in the order of 10-8 M. The internal Gpm1 sequence motifs, directly interacting with VTR (aa 116-158) and FN (aa 138-175) were mapped using chemical crosslinking and mass spectrometry. Synthetic peptides with matching sequences significantly inhibited formation of the Gpm1-VTR and Gpm1-FN complexes. A molecular model of the Gpm1-VTR complex was developed. These results provide the first structural insights into the adhesin function of candidal surface-exposed Gpm1.


Asunto(s)
Candida albicans/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas Fúngicas/metabolismo , Fosfoglicerato Mutasa/metabolismo , Candida albicans/química , Membrana Celular/metabolismo , Pared Celular/metabolismo , Proteínas de la Matriz Extracelular/química , Fibronectinas/química , Fibronectinas/metabolismo , Proteínas Fúngicas/química , Humanos , Modelos Moleculares , Fosfoglicerato Mutasa/química , Unión Proteica , Resonancia por Plasmón de Superficie/métodos , Factores de Virulencia/metabolismo , Vitronectina/química , Vitronectina/metabolismo
15.
Proteins ; 78(7): 1691-704, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20143318

RESUMEN

Phosphoglycerate mutase (PGM) and enolase are consecutive enzymes in the glycolytic pathway. We used molecular dynamics simulation to examine the interaction of human B-type PGM (dPGM-B) and neuron-specific enolase (NSE). Specifically, we studied the interactions of 31 orientations of these enzymes by means of the effective energy function implicit solvation method. Interactions between active regions of the enzymes occurred preferentially, although the strongest interactions appeared to be between the back side of NSE and the active regions of dPGM-B. Cleavage of 2PG from dPGM-B was investigated, and the Ser(14)-Leu(30) loop of dPGM-B is suggested as a cleavage site and, likely, another entrance site of a ligand. Substrate channeling between the enzymes was observed when NSE with its active regions Leu(11)-Asn(16), Arg(49)-Lys(59), and Gly(155)-Ala(158) covered the Ser(14)-Leu(30) loop of dPGM-B. Analyses of the results make us believe that the channeling between PGM and enolase "benefits" from weak interaction. The probability of formation of channeling favorable complex is estimated to be up to 5%, while functional interaction between NSE and dPGM-B might be as high as 20%. NSE and dPGM-B functional interaction seems not to be isotype specific.


Asunto(s)
Simulación de Dinámica Molecular , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Fosfopiruvato Hidratasa/química , Fosfopiruvato Hidratasa/metabolismo , Humanos , Isoenzimas , Unión Proteica , Mapeo de Interacción de Proteínas , Termodinámica
16.
ACS Chem Biol ; 15(3): 632-639, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32069008

RESUMEN

Post-translational modifications play vital roles in fine-tuning a myriad of physiological processes, and one of the most important modifications is acetylation. Here, we report a ligand-directed site-selective acetylation using KHAc, a derivative of a phosphoglycerate mutase 1 (PGAM1) inhibitor. KHAc binds to PGAM1 and transfers its acetyl group to the ε-NH2 of Lys100 to inactivate the enzyme. The acetyl transfer process was visualized by time-resolved crystallography, demonstrating that the transfer is driven by proximity effects. KHAc was capable of selectively and effectively acetylating Lys100 of PGAM1 in cultured human cells, accompanied by inhibited F-actin formation. Similar strategies could be used for exogenous control of other lysine post-translational modifications.


Asunto(s)
Inhibidores Enzimáticos/química , Compuestos Heterocíclicos/química , Fosfoglicerato Mutasa/química , Acetilación , Actinas/metabolismo , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Cristalización , Células HEK293 , Humanos , Ligandos , Mutación , Fosfoglicerato Mutasa/antagonistas & inhibidores , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional
17.
Biochim Biophys Acta ; 1784(1): 100-5, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17728195

RESUMEN

Histidine phosphorylation is important in prokaryotes and occurs to the extent of 6% of total phosphorylation in eukaryotes. Nevertheless phosphohistidine residues are not normally observed in proteins due to rapid hydrolysis of the phosphoryl group under acidic conditions. Many rapid processes employ phosphohistidines, including the bacterial phosphoenolpyruvate:sugar phosphotransferase system (PTS), the bacterial two-component systems and reactions catalyzed by enzymes such as nucleoside diphosphate kinase and succinyl-CoA synthetase. In the PTS, the NMR structure of the phosphohistidine moiety of the phosphohistidine-containing protein was determined but no X-ray structures of phosphohistidine forms of PTS proteins have been elucidated. There have been crystal structures of a few phosphohistidine-containing proteins determined: nucleoside diphosphate kinase, succinyl-CoA synthetase, a cofactor-dependent phosphoglycerate mutase and the protein PAE2307 from the hyperthermophilic archaeon Pyrobaculum aerophilum. A common theme for these stable phosphohistidines is the occurrence of ion-pair hydrogen bonds (salt bridges) involving the non-phosphorylated nitrogen atom of the histidine imidazole ring with an acidic amino acid side chain.


Asunto(s)
Histidina/análogos & derivados , Histidina/metabolismo , Cristalografía por Rayos X , Histidina/química , Enlace de Hidrógeno , Nucleósido-Difosfato Quinasa/química , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/química , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Fosfoglicerato Mutasa/química , Fosforilación , Succinato-CoA Ligasas/química , Succinato-CoA Ligasas/metabolismo
18.
Methods Enzymol ; 422: 288-304, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17628145

RESUMEN

Escherichia coli protein SixA was the first identified histidine protein phosphatase that dephosphorylates the histidine-containing phosphotransfer (HPt) domain of histidine kinase ArcB. The crystal structures of the free and tungstate-bound forms of SixA revealed an alpha/beta architecture with a fold unlike those previously described in eukaryotic protein phosphatases, but related to a family of phosphatases containing the arginine-histidine-glycine (RHG) motif at their active sites. Compared with these RHG phosphatases, SixA lacks an extra alpha-helical subdomain that forms a lid over the active site, thereby forming a relatively shallow groove important for accommodating the kidney-shaped four-helix bundle of the HPt domain. Sequence database searches revealed that a single SixA homolog was found in a variety of bacteria, where two homologs were found in some bacteria while no homolog was found in others. No SixA homologs were found in the majority of firmicutes and euryarchaea. Structure-based examination and multiple alignment of sequences revealed SixA active residues from loop beta1-H2, which might assist in the identification of SixA homologs among RHG phosphatases even with poor amino acid identity.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Proteínas de la Membrana/metabolismo , Fosfoproteínas Fosfatasas/química , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Histidina/metabolismo , Proteínas de la Membrana/química , Modelos Moleculares , Datos de Secuencia Molecular , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Proteínas Quinasas/química
19.
Mol Biochem Parasitol ; 156(2): 210-6, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17897734

RESUMEN

Phosphoglycerate mutase (PGM, EC 5.4.2.1) catalyzes the isomerization of 3-phosphoglycerate and 2-phosphoglycerate in glycolysis and gluconeogenesis. Two distinct types of PGM exist in nature, one that requires 2,3-bisphosphoglycerate as a cofactor (dPGM) and another that does not (iPGM). The two enzymes are structurally distinct and possess different mechanisms of action. In any particular organism, one form may exist or both. Nematodes possess the iPGM form whereas mammals have dPGM. In the present study, we have cloned and expressed iPGM from Onchocerca volvulus and described the catalytic properties of O. volvulus, Brugia malayi and Caenorhabditis elegans iPGM enzymes. Temperature and pH optima were determined for each enzyme. Like other iPGM enzymes, the activities of the nematode iPGM enzymes were dependent on the presence of divalent ions. Inactivation by EDTA could be restored most effectively by magnesium and manganese ions. Kinetic parameters and specific activities of the various recombinant enzymes were determined. The high similarity in catalytic properties among the enzymes indicates that a single enzyme inhibitor would likely be effective against all nematode enzymes. Inhibition of iPGM activity in vivo may lead to lethality as indicated by RNAi studies in C. elegans. Our results support the development of iPGM as a promising drug target in parasitic nematodes.


Asunto(s)
Brugia Malayi/enzimología , Caenorhabditis elegans/enzimología , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Onchocerca volvulus/enzimología , Fosfoglicerato Mutasa/genética , Fosfoglicerato Mutasa/metabolismo , Animales , Brugia Malayi/genética , Caenorhabditis elegans/genética , Cationes Bivalentes/farmacología , Clonación Molecular , Coenzimas/farmacología , Ácido Edético/farmacología , Inhibidores Enzimáticos/farmacología , Estabilidad de Enzimas , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , Onchocerca volvulus/genética , Fosfoglicerato Mutasa/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Temperatura
20.
Mol Biosyst ; 3(7): 495-506, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17579775

RESUMEN

We recently reported the pharmacological screening of a natural products-inspired library of spiroepoxide probes, resulting in the discovery of an agent MJE3 that displayed anti-proliferative effects in human breast cancer cells. MJE3 was found to covalently inactivate phosphoglycerate mutase-1 (PGAM1), a glycolytic enzyme with postulated roles in cancer cell metabolism and proliferation. Considering that MJE3 is one of the first examples of a cell-permeable, small-molecule inhibitor for PGAM1, we pursued a detailed examination of its mechanism and structural requirements for covalent inactivation. MJE3 was found to label PGAM1 on lysine-100, a conserved active site residue implicated in substrate recognition. Structural features of MJE3 important for PGAM1 labeling included two key recognition elements (an indole ring and carboxylic acid), the stereochemical orientation of the spiroepoxide, and presentation of these various binding/reactive groups on a rigid cyclohexane scaffold. Modeling studies of the docked MJE3-PGAM1 complex provide a structural rationale for these stringent requirements. Overall, these studies indicate that a special combination of binding and reactive elements are united in the MJE3 structure to inactivate PGAM1. More generally, our findings provide further evidence that useful pharmacological tools can emerge from screening structurally diverse libraries of protein-reactive probes.


Asunto(s)
Compuestos Epoxi/química , Fosfoglicerato Mutasa/química , Animales , Sitios de Unión/genética , Western Blotting , Células COS , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Compuestos Epoxi/síntesis química , Compuestos Epoxi/farmacología , Humanos , Espectrometría de Masas , Modelos Moleculares , Estructura Molecular , Mutación , Fosfoglicerato Mutasa/genética , Fosfoglicerato Mutasa/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Relación Estructura-Actividad , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA