Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell ; 187(5): 1296-1311.e26, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38428397

RESUMEN

Most membrane proteins are modified by covalent addition of complex sugars through N- and O-glycosylation. Unlike proteins, glycans do not typically adopt specific secondary structures and remain very mobile, shielding potentially large fractions of protein surface. High glycan conformational freedom hinders complete structural elucidation of glycoproteins. Computer simulations may be used to model glycosylated proteins but require hundreds of thousands of computing hours on supercomputers, thus limiting routine use. Here, we describe GlycoSHIELD, a reductionist method that can be implemented on personal computers to graft realistic ensembles of glycan conformers onto static protein structures in minutes. Using molecular dynamics simulation, small-angle X-ray scattering, cryoelectron microscopy, and mass spectrometry, we show that this open-access toolkit provides enhanced models of glycoprotein structures. Focusing on N-cadherin, human coronavirus spike proteins, and gamma-aminobutyric acid receptors, we show that GlycoSHIELD can shed light on the impact of glycans on the conformation and activity of complex glycoproteins.


Asunto(s)
Glicoproteínas , Simulación de Dinámica Molecular , Humanos , Microscopía por Crioelectrón , Glicoproteínas/química , Glicosilación , Polisacáridos/química
2.
Nat Methods ; 18(4): 366-368, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33828273

RESUMEN

We are at the beginning of a genomic revolution in which all known species are planned to be sequenced. Accessing such data for comparative analyses is crucial in this new age of data-driven biology. Here, we introduce an improved version of DIAMOND that greatly exceeds previous search performances and harnesses supercomputing to perform tree-of-life scale protein alignments in hours, while matching the sensitivity of the gold standard BLASTP.


Asunto(s)
Biología Computacional/métodos , Proteínas/química , Alineación de Secuencia , Algoritmos
3.
J Chem Phys ; 157(17): 174801, 2022 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-36347673

RESUMEN

Lipid membranes are integral building blocks of living cells and perform a multitude of biological functions. Currently, molecular simulations of cellular-scale membrane remodeling processes at atomic resolution are extremely difficult, due to their size, complexity, and the large times-scales on which these processes occur. Instead, elastic membrane models are used to simulate membrane shapes and transitions between them and to infer their properties and functions. Unfortunately, an efficiently parallelized open-source simulation code to do so has been lacking. Here, we present TriMem, a parallel hybrid Monte Carlo simulation engine for triangulated lipid membranes. The kernels are efficiently coded in C++ and wrapped with Python for ease-of-use. The parallel implementation of the energy and gradient calculations and of Monte Carlo flip moves of edges in the triangulated membrane enable us to simulate large and highly curved membrane structures. For validation, we reproduce phase diagrams of vesicles with varying surface-to-volume ratios and area difference. We also compute the density of states to verify correct Boltzmann sampling. The software can be used to tackle a range of large-scale membrane remodeling processes as a step toward cell-scale simulations. Additionally, extensive documentation make the software accessible to the broad biophysics and computational cell biology communities.


Asunto(s)
Lípidos , Programas Informáticos , Método de Montecarlo , Simulación por Computador
4.
Angew Chem Int Ed Engl ; 60(43): 23419-23426, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34387025

RESUMEN

Mechanistic insights into protein-ligand interactions can yield chemical tools for modulating protein function and enable their use for therapeutic purposes. For the homodimeric enzyme tRNA-guanine transglycosylase (TGT), a putative virulence target of shigellosis, ligand binding has been shown by crystallography to transform the functional dimer geometry into an incompetent twisted one. However, crystallographic observation of both end states does neither verify the ligand-induced transformation of one dimer into the other in solution nor does it shed light on the underlying transformation mechanism. We addressed these questions in an approach that combines site-directed spin labeling (SDSL) with distance measurements based on pulsed electron-electron double resonance (PELDOR or DEER) spectroscopy. We observed an equilibrium between the functional and twisted dimer that depends on the type of ligand, with a pyranose-substituted ligand being the most potent one in shifting the equilibrium toward the twisted dimer. Our experiments suggest a dissociation-association mechanism for the formation of the twisted dimer upon ligand binding.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pentosiltransferasa/metabolismo , Quinazolinonas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Simulación por Computador , Espectroscopía de Resonancia por Spin del Electrón , Ligandos , Mutación , Pentosiltransferasa/química , Pentosiltransferasa/genética , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Quinazolinonas/química , Zymomonas/enzimología
5.
Biochemistry ; 57(26): 3953-3965, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29862811

RESUMEN

The bacterial enzyme tRNA-guanine transglycosylase (TGT) is involved in the biosynthesis of queuosine, a modified nucleoside present in the anticodon wobble position of tRNAHis, tRNATyr, tRNAAsp, and tRNAAsn. Although it forms a stable homodimer endowed with two active sites, it is, for steric reasons, able to bind and convert only one tRNA molecule at a time. In contrast, its mammalian counterpart constitutes a heterodimer consisting of a catalytic and a noncatalytic subunit, termed QTRT1 and QTRT2, respectively. Both subunits are homologous to the bacterial enzyme, yet only QTRT1 possesses all the residues required for substrate binding and catalysis. In mice, genetic inactivation of the TGT results in the uncontrolled oxidation of tetrahydrobiopterin and, accordingly, phenylketonuria-like symptoms. For this reason and because of the recent finding that mammalian TGT may be utilized for the treatment of multiple sclerosis, this enzyme is of potential medical relevance, rendering detailed knowledge of its biochemistry and structural architecture highly desirable. In this study, we performed the kinetic characterization of the murine enzyme, investigated potential quaternary structures of QTRT1 and QTRT2 via noncovalent mass spectrometry, and, finally, determined the crystal structure of the murine noncatalytic TGT subunit, QTRT2. In the crystal, QTRT2 is clearly present as a homodimer that is strikingly similar to that formed by bacterial TGT. In particular, a cluster of four aromatic residues within the interface of the bacterial TGT, which constitutes a "hot spot" for dimer stability, is present in a similar constellation in QTRT2.


Asunto(s)
Pentosiltransferasa/química , Multimerización de Proteína , Subunidades de Proteína/química , Animales , Cinética , Ratones , Estructura Cuaternaria de Proteína
6.
Angew Chem Int Ed Engl ; 57(32): 10085-10090, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29927035

RESUMEN

The enzyme tRNA-guanine transglycosylase, a target to fight Shigellosis, recognizes tRNA only as a homodimer and performs full nucleobase exchange at the wobble position. Active-site inhibitors block the enzyme function by competitively replacing tRNA. In solution, the wild-type homodimer dissociates only marginally, whereas mutated variants show substantial monomerization in solution. Surprisingly, one inhibitor transforms the protein into a twisted state, whereby one monomer unit rotates by approximately 130°. In this altered geometry, the enzyme is no longer capable of binding and processing tRNA. Three sugar-type inhibitors have been designed and synthesized, which bind to the protein in either the functionally competent or twisted inactive state. They crystallize with the enzyme side-by-side under identical conditions from the same crystallization well. Possibly, the twisted inactive form corresponds to a resting state of the enzyme, important for its functional regulation.


Asunto(s)
Pentosiltransferasa/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Modelos Moleculares , Estructura Molecular , Pentosiltransferasa/antagonistas & inhibidores , Pentosiltransferasa/química
7.
Proteins ; 82(9): 2041-53, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24623648

RESUMEN

Clostridium propionicum is the only organism known to ferment ß-alanine, a constituent of coenzyme A (CoA) and the phosphopantetheinyl prosthetic group of holo-acyl carrier protein. The first step in the fermentation is a CoA-transfer to ß-alanine. Subsequently, the resulting ß-alanyl-CoA is deaminated by the enzyme ß-alanyl-CoA:ammonia lyase (Acl) to reversibly form ammonia and acrylyl-CoA. We have determined the crystal structure of Acl in its apo-form at a resolution of 0.97 Å as well as in complex with CoA at a resolution of 1.59 Å. The structures reveal that the enyzme belongs to a superfamily of proteins exhibiting a so called "hot dog fold" which is characterized by a five-stranded antiparallel ß-sheet with a long α-helix packed against it. The functional unit of all "hot dog fold" proteins is a homodimer containing two equivalent substrate binding sites which are established by the dimer interface. In the case of Acl, three functional dimers combine to a homohexamer strongly resembling the homohexamer formed by YciA-like acyl-CoA thioesterases. Here, we propose an enzymatic mechanism based on the crystal structure of the Acl·CoA complex and molecular docking.


Asunto(s)
Amoníaco-Liasas/ultraestructura , Clostridium/enzimología , Pliegue de Proteína , Secuencia de Aminoácidos , Sitios de Unión , Coenzima A/metabolismo , Cristalografía por Rayos X , Fermentación/fisiología , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Complejos Multiproteicos , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Alineación de Secuencia , beta-Alanina/metabolismo
8.
Proteins ; 82(10): 2713-32, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24975703

RESUMEN

Interference with protein-protein interactions of interfaces larger than 1500 Ų by small drug-like molecules is notoriously difficult, particularly if targeting homodimers. The tRNA modifying enzyme Tgt is only functionally active as a homodimer. Thus, blocking Tgt dimerization is a promising strategy for drug therapy as this protein is key to the development of Shigellosis. Our goal was to identify hot-spot residues which, upon mutation, result in a predominantly monomeric state of Tgt. The detailed understanding of the spatial location and stability contribution of the individual interaction hot-spot residues and the plasticity of motifs involved in the interface formation is a crucial prerequisite for the rational identification of drug-like inhibitors addressing the respective dimerization interface. Using computational analyses, we identified hot-spot residues that contribute particularly to dimer stability: a cluster of hydrophobic and aromatic residues as well as several salt bridges. This in silico prediction led to the identification of a promising double mutant, which was validated experimentally. Native nano-ESI mass spectrometry showed that the dimerization of the suggested mutant is largely prevented resulting in a predominantly monomeric state. Crystal structure analysis and enzyme kinetics of the mutant variant further support the evidence for enhanced monomerization and provide first insights into the structural consequences of the dimer destabilization.


Asunto(s)
Modelos Moleculares , Proteínas Mutantes/química , Pentosiltransferasa/química , ARN de Transferencia/metabolismo , Sustitución de Aminoácidos , Proteínas Arqueales/química , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biología Computacional , Bases de Datos de Proteínas , Dimerización , Estabilidad de Enzimas , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Sistemas Especialistas , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/metabolismo , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
9.
ACS Omega ; 8(48): 46051-46065, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38075755

RESUMEN

The Shigella pathogenicity factor IpgC belongs to the class II of type III secretion system chaperones, whose members are characterized by a tetratricopeptide repeat (TPR) domain consisting of three and a half TPR motifs. Since IpgC is essential for Shigella virulence, we determined a high-resolution crystal structure of this chaperone to facilitate its use as a target for the structure-based design of anti-shigellosis compounds. The crystal structure revealed two possible homodimer assemblies, which strongly differ from the homodimer architectures so far known for IpgC and orthologues thereof. Through crystallographic fragment screening, we identified 10 small molecules that bind to IpgC and, therefore, are available for expansion to generate larger, more potent binders. A follow-up compound, based on one of our fragment hits, binds to a strictly conserved site, which overlaps with the binding site of the chaperone's substrates, IpaB and IpaC. Therefore, it constitutes a promising starting point for the design of functional IpgC inhibitors.

10.
ACS Chem Biol ; 17(8): 2229-2247, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35815944

RESUMEN

In tRNAAsp, tRNAAsn, tRNATyr, and tRNAHis of most bacteria and eukaryotes, the anticodon wobble position may be occupied by the modified nucleoside queuosine, which affects the speed and the accuracy of translation. Since eukaryotes are not able to synthesize queuosine de novo, they have to salvage queuine (the queuosine base) as a micronutrient from food and/or the gut microbiome. The heterodimeric Zn2+ containing enzyme tRNA-guanine transglycosylase (TGT) catalyzes the insertion of queuine into the above-named tRNAs in exchange for the genetically encoded guanine. This enzyme has attracted medical interest since it was shown to be potentially useful for the treatment of multiple sclerosis. In addition, TGT inactivation via gene knockout leads to the suppressed cell proliferation and migration of certain breast cancer cells, which may render this enzyme a potential target for the design of compounds supporting breast cancer therapy. As a prerequisite to fully exploit the medical potential of eukaryotic TGT, we have determined and analyzed a number of crystal structures of the functional murine TGT with and without bound queuine. In addition, we have investigated the importance of two residues of its non-catalytic subunit on dimer stability and determined the Michaelis-Menten parameters of murine TGT with respect to tRNA and several natural and artificial nucleobase substrates. Ultimately, on the basis of available TGT crystal structures, we provide an entirely conclusive reaction mechanism for this enzyme, which in detail explains why the TGT-catalyzed insertion of some nucleobases into tRNA occurs reversibly while that of others is irreversible.


Asunto(s)
Pentosiltransferasa/química , Animales , Células Eucariotas/metabolismo , Femenino , Guanina/metabolismo , Humanos , Ratones , Nucleósido Q , ARN de Transferencia/química
11.
ACS Chem Biol ; 17(7): 1745-1755, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35763700

RESUMEN

Understanding the structural arrangements of protein oligomers can support the design of ligands that interfere with their function in order to develop new therapeutic concepts for disease treatment. Recent crystallographic studies have elucidated a novel twisted and functionally inactive form of the homodimeric enzyme tRNA-guanine transglycosylase (TGT), a putative target in the fight against shigellosis. Active-site ligands have been identified that stimulate the rearrangement of one monomeric subunit by 130° against the other one to form an inactive twisted homodimer state. To assess whether the crystallographic observations also reflect the conformation in solution and rule out effects from crystal packing, we performed 19F-NMR spectroscopy with the introduction of 5-fluorotryptophans at four sites in TGT. The inhibitor-induced conformation of TGT in solution was assessed based on 19F-NMR chemical shift perturbations. We investigated the effect of C(4) substituted lin-benzoguanine ligands and identified a correlation between dynamic protein rearrangements and ligand-binding features in the corresponding crystal structures. These involve the destabilization of a helix next to the active site and the integrity of a flexible loop-helix motif. Ligands that either completely lack an attached C(4) substituent or use it to stabilize the geometry of the functionally competent dimer state do not indicate the presence of the twisted dimer form in the NMR spectra. The perturbation of crucial structural motifs in the inhibitors correlates with an increasing formation of the inactive twisted dimer state, suggesting these ligands are able to shift a conformational equilibrium from active C2-symmetric to inactive twisted dimer conformations. These findings suggest a novel concept for the design of drug candidates for further development.


Asunto(s)
Zymomonas , Dominio Catalítico , Cristalografía por Rayos X , Guanina/metabolismo , Ligandos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Pentosiltransferasa/química , Conformación Proteica , ARN de Transferencia/química , Zymomonas/química
12.
ACS Chem Biol ; 16(6): 1090-1098, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34081441

RESUMEN

Interference with protein-protein interfaces represents an attractive as well as challenging option for therapeutic intervention and drug design. The enzyme tRNA-guanine transglycosylase, a target to fight Shigellosis, is only functional as a homodimer. Although we previously produced monomeric variants by site-directed mutagenesis, we only crystallized the functional dimer, simply because upon crystallization the local protein concentration increases and favors formation of the dimer interface, which represents an optimal and highly stable packing of the protein in the solid state. Unfortunately, this prevents access to structural information about the interface geometry in its monomeric state and complicates the development of modulators that can interfere with and prevent dimer formation. Here, we report on a cysteine-containing protein variant in which, under oxidizing conditions, a disulfide linkage is formed. This reinforces a novel packing geometry of the enzyme. In this captured quasi-monomeric state, the monomer units arrange in a completely different way and, thus, expose a loop-helix motif, originally embedded into the old interface, now to the surface. The motif adopts a geometry incompatible with the original dimer formation. Via the soaking of fragments into the crystals, we identified several hits accommodating a cryptic binding site next to the loop-helix motif and modulated its structural features. Our study demonstrates the druggability of the interface by breaking up the homodimeric protein using an introduced disulfide cross-link. By rational concepts, we increased the potency of these fragments to a level where we confirmed their binding by NMR to a nondisulfide-linked TGT variant. The idea of intermediately introducing a disulfide linkage may serve as a general concept of how to transform a homodimer interface into a quasi-monomeric state and give access to essential structural and design information.


Asunto(s)
Disulfuros/química , Pentosiltransferasa/química , Bibliotecas de Moléculas Pequeñas/farmacología , Zymomonas/enzimología , Sitios de Unión/efectos de los fármacos , Ligandos , Modelos Moleculares , Multimerización de Proteína/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Zymomonas/química
13.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 6): 685-97, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20516621

RESUMEN

Most commercially available crystallization screens are sparse-matrix screens with a predominance of inorganic salts and polyethylene glycols (PEGs) as precipitants. It was noted that commercially available screens are largely unsatisfactory for the purpose of the crystallization of multimeric protein and protein-nucleic acid complexes. This was reasoned to be a consequence of the redundancy in screening crystallization parameter space by the predominance of PEG as a precipitant in standard screens and it was suggested that this limitation could be overcome by introducing a variety of other organic polymers. Here, a set of 288 crystallization conditions was devised based on alternative polymeric precipitants and tested against a set of 20 different proteins/complexes; finally, a screen comprising the 96 most promising conditions designed to complement PEG- and salt-based commercial screens was proposed.


Asunto(s)
Cristalización/métodos , Polietilenglicoles/química , Proteínas/análisis , Animales , Cristalografía por Rayos X , Humanos , Concentración de Iones de Hidrógeno , Proteínas/química , Sales (Química)/química
14.
ACS Chem Biol ; 15(11): 3021-3029, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33166460

RESUMEN

Bacterial tRNA-guanine transglycosylase (Tgt) is involved in the biosynthesis of the modified tRNA nucleoside queuosine present in the anticodon wobble position of tRNAs specific for aspartate, asparagine, histidine, and tyrosine. Inactivation of the tgt gene leads to decreased pathogenicity of Shigella bacteria. Therefore, Tgt constitutes a putative target for Shigellosis drug therapy. Since it is only active as homodimer, interference with dimer-interface formation may, in addition to active-site inhibition, provide further means to disable this protein. A cluster of four aromatic residues seems important to stabilize the homodimer. We mutated residues of this aromatic cluster and analyzed each mutated variant with respect to the dimer and thermal stability or enzyme activity by applying native mass spectrometry, a thermal shift assay, enzyme kinetics, and X-ray crystallography. Our structural studies indicate a strong influence of pH on the homodimer stability. Apparently, protonation of a histidine within the aromatic cluster supports the collapse of an essential structural motif within the dimer interface at slightly acidic pH.


Asunto(s)
Pentosiltransferasa/química , Zymomonas/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Estabilidad de Enzimas , Modelos Moleculares , Mutación , Pentosiltransferasa/genética , Conformación Proteica , Multimerización de Proteína , Zymomonas/química , Zymomonas/genética
15.
J Med Chem ; 63(13): 6802-6820, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32515955

RESUMEN

Fragment-based lead discovery was applied to tRNA-guanine transglycosylase, an enzyme modifying post-transcriptionally tRNAs in Shigella, the causative agent of shigellosis. TGT inhibition prevents translation of Shigella's virulence factor VirF, hence reducing pathogenicity. One discovered fragment opens a transient subpocket in the preQ1-recognition site by pushing back an aspartate residue. This step is associated with reorganization of further amino acids structurally transforming a loop adjacent to the recognition site by duplicating the volume of the preQ1-recognition pocket. We synthesized 6-carboxamido-, 6-hydrazido-, and 4-guanidino-benzimidazoles to target the opened pocket, including a dihydro-imidazoquinazoline with a propyn-1-yl exit vector pointing into the transient pocket and displacing a conserved water network. MD simulations and hydration-site analysis suggest water displacement to contribute favorably to ligand binding. A cysteine residue, exclusively present in bacterial TGTs, serves as gatekeeper of the transient subpocket. It becomes accessible upon pocket opening for selective covalent attachment of electrophilic ligands in eubacterial TGTs.


Asunto(s)
Pentosiltransferasa/metabolismo , Bencimidazoles/farmacología , Sitios de Unión , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Ligandos , Modelos Moleculares , Pentosiltransferasa/química , Conformación Proteica , Shigella/enzimología
16.
J Chem Theory Comput ; 15(5): 3390-3401, 2019 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-30939006

RESUMEN

Ensemble refinement produces structural ensembles of flexible and dynamic biomolecules by integrating experimental data and molecular simulations. Here we present two efficient numerical methods to solve the computationally challenging maximum-entropy problem arising from a Bayesian formulation of ensemble refinement. Recasting the resulting constrained weight optimization problem into an unconstrained form enables the use of gradient-based algorithms. In two complementary formulations that differ in their dimensionality, we optimize either the log-weights directly or the generalized forces appearing in the explicit analytical form of the solution. We first demonstrate the robustness, accuracy, and efficiency of the two methods using synthetic data. We then use NMR J-couplings to reweight an all-atom molecular dynamics simulation ensemble of the disordered peptide Ala-5 simulated with the AMBER99SB*-ildn-q force field. After reweighting, we find a consistent increase in the population of the polyproline-II conformations and a decrease of α-helical-like conformations. Ensemble refinement makes it possible to infer detailed structural models for biomolecules exhibiting significant dynamics, such as intrinsically disordered proteins, by combining input from experiment and simulation in a balanced manner.


Asunto(s)
Algoritmos , Simulación de Dinámica Molecular , Péptidos/química , Resonancia Magnética Nuclear Biomolecular
17.
PLoS One ; 12(4): e0175723, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28419165

RESUMEN

For the efficient pathogenesis of Shigella, the causative agent of bacillary dysentery, full functionality of tRNA-guanine transglycosylase (TGT) is mandatory. TGT performs post-transcriptional modifications of tRNAs in the anticodon loop taking impact on virulence development. This suggests TGT as a putative target for selective anti-shigellosis drug therapy. Since bacterial TGT is only functional as homodimer, its activity can be inhibited either by blocking its active site or by preventing dimerization. Recently, we discovered that in some crystal structures obtained by soaking the full conformational adaptation most likely induced in solution upon ligand binding is not displayed. Thus, soaked structures may be misleading and suggest irrelevant binding modes. Accordingly, we re-investigated these complexes by co-crystallization. The obtained structures revealed large conformational rearrangements not visible in the soaked complexes. They result from spatial perturbations in the ribose-34/phosphate-35 recognition pocket and, consequently, an extended loop-helix motif required to prevent access of water molecules into the dimer interface loses its geometric integrity. Thermodynamic profiles of ligand binding in solution indicate favorable entropic contributions to complex formation when large conformational adaptations in the dimer interface are involved. Native MS titration experiments reveal the extent to which the homodimer is destabilized in the presence of each inhibitor. Unexpectedly, one ligand causes a complete rearrangement of subunit packing within the homodimer, never observed in any other TGT crystal structure before. Likely, this novel twisted dimer is catalytically inactive and, therefore, suggests that stabilizing this non-productive subunit arrangement may be used as a further strategy for TGT inhibition.


Asunto(s)
Proteínas Bacterianas/química , Modelos Moleculares , Multimerización de Proteína , ARN de Transferencia/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalización , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Pentosiltransferasa/antagonistas & inhibidores , Pentosiltransferasa/química , Pentosiltransferasa/metabolismo , Unión Proteica , Conformación Proteica , Dominios Proteicos , Estabilidad Proteica , Estructura Secundaria de Proteína , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Soluciones , Termodinámica , Zymomonas/enzimología
18.
J Mol Biol ; 342(5): 1613-24, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15364585

RESUMEN

In vivo studies with the fruit-fly Drosophila melanogaster have shown that the Sniffer protein prevents age-dependent and oxidative stress-induced neurodegenerative processes. Sniffer is a NADPH-dependent carbonyl reductase belonging to the enzyme family of short-chain dehydrogenases/reductases (SDRs). The crystal structure of the homodimeric Sniffer protein from Drosophila melanogaster in complex with NADP+ has been determined by multiple-wavelength anomalous dispersion and refined to a resolution of 1.75 A. The observed fold represents a typical dinucleotide-binding domain as detected for other SDRs. With respect to the cofactor-binding site and the region referred to as substrate-binding loop, the Sniffer protein shows a striking similarity to the porcine carbonyl reductase (PTCR). This loop, in both Sniffer and PTCR, is substantially shortened compared to other SDRs. In most enzymes of the SDR family this loop adopts a well-defined conformation only after substrate binding and remains disordered in the absence of any bound ligands or even if only the dinucleotide cofactor is bound. In the structure of the Sniffer protein, however, the conformation of this loop is well defined, although no substrate is present. Molecular modeling studies provide an idea of how binding of substrate molecules to Sniffer could possibly occur.


Asunto(s)
Oxidorreductasas de Alcohol/química , Proteínas de Drosophila/química , Drosophila melanogaster/enzimología , NADP/metabolismo , Fármacos Neuroprotectores/química , Oxidorreductasas de Alcohol/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Proteínas de Drosophila/metabolismo , Humanos , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , NADP/química , Fármacos Neuroprotectores/metabolismo , Unión Proteica , Conformación Proteica , Homología de Secuencia de Aminoácido , Porcinos
19.
J Mol Biol ; 338(1): 55-75, 2004 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-15050823

RESUMEN

The enzyme tRNA-guanine transglycosylase (TGT) is involved in the pathogenicity of Shigellae. As the crystal structure of this protein is known, it is a putative target for the structure-based design of inhibitors. Here we report a crystallographic study of several new ligands exhibiting a 2,6-diamino-3H-quinazolin-4-one scaffold, which has been shown recently to be a promising template for TGT-inhibitors. Crystal structure analysis of these complexes has revealed an unexpected movement of the side-chain of Asp102. A detailed analysis of the water network disrupted by this rotation has lead to the derivation of a new composite pharmacophore. A virtual screening has been performed based on this pharmacophore hypothesis and several new inhibitors of micromolar binding affinity with new skeletons have been discovered.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Pentosiltransferasa/química , Quinazolinas/química , Quinazolinas/síntesis química , Zymomonas/enzimología , Ácido Aspártico/metabolismo , Sitios de Unión , Técnicas Químicas Combinatorias , Cristalografía por Rayos X , Bases de Datos Factuales , Diseño de Fármacos , Inhibidores Enzimáticos/química , Ligandos , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Pentosiltransferasa/metabolismo , Relación Estructura-Actividad , Zymomonas/química
20.
J Mol Biol ; 328(3): 609-21, 2003 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-12706720

RESUMEN

Following acetate, propionate is the second most abundant low molecular mass carbon compound found in soil. Many microorganisms, including most, if not all fungi, as well as several aerobic bacteria, such as Escherichia coli and Salmonella enterica oxidize propionate via the methylcitrate cycle. The enzyme 2-methylisocitrate lyase (PrpB) from Escherichia coli catalysing the last step of this cycle, the cleavage of 2-methylisocitrate to pyruvate and succinate, was crystallised and its structure determined to a resolution of 1.9A. The enzyme, which strictly depends on Mg(2+) for catalysis, belongs to the isocitrate lyase protein family. A common feature of members of this enzyme family is the movement of a so-called "active site loop" from an open into a closed conformation upon substrate binding thus shielding the reactants from the surrounding solvent. Since in the presented structure, PrpB contains, apart from a Mg(2+), no ligand, the active site loop is found in an open conformation. This conformation, however, differs significantly from the open conformation present in the so far known structures of ligand-free isocitrate lyases. A possible impact of this observation with respect to the different responses of isocitrate lyases and PrpB upon treatment with the common inhibitor 3-bromopyruvate is discussed. Based on the structure of ligand-bound isocitrate lyase from Mycobacterium tuberculosis a model of the substrate-bound PrpB enzyme in its closed conformation was created which provides hints towards the substrate specificity of this enzyme.


Asunto(s)
Liasas de Carbono-Carbono/química , Liasas de Carbono-Carbono/metabolismo , Escherichia coli/enzimología , Modelos Moleculares , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Isocitratoliasa/química , Ligandos , Modelos Químicos , Datos de Secuencia Molecular , Propionatos/metabolismo , Conformación Proteica , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA