Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Molecules ; 27(18)2022 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-36144626

RESUMEN

Previous investigations of the Leishmania infantum eIF4A-like protein (LieIF4A) as a potential drug target delivered cholestanol derivatives inhibitors. Here, we investigated the mode of action of cholesterol derivatives as a novel scaffold structure of LieIF4A inhibitors on the RNA-dependent ATPase activity of LieIF4A and its mammalian ortholog (eIF4AI). We compared their biochemical effects on RNA-dependent ATPase activities of both proteins and investigated if rocaglamide, a known inhibitor of eIF4A, could affect LieIF4A as well. Kinetic measurements were conducted at different concentrations of ATP, of the compound and in the presence of saturating whole yeast RNA concentrations. Kinetic analyses showed different ATP binding affinities for the two enzymes as well as different sensitivities to 7-α-aminocholesterol and rocaglamide. The 7-α-aminocholesterol inhibited LieIF4A with a higher binding affinity relative to cholestanol analogs. Cholesterol, another tested sterol, had no effect on the ATPase activity of LieIF4A or eIF4AI. The 7-α-aminocholesterol demonstrated an anti-Leishmania activity on L. infantum promastigotes. Additionally, docking simulations explained the importance of the double bond between C5 and C6 in 7-α-aminocholesterol and the amino group in the C7 position. In conclusion, Leishmania and mammalian eIF4A proteins appeared to interact differently with effectors, thus making LieIF4A a potential drug against leishmaniases.


Asunto(s)
Factor 4A Eucariótico de Iniciación , Leishmania infantum , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Colestanoles/metabolismo , Colesterol/metabolismo , Factor 4A Eucariótico de Iniciación/química , Factor 4A Eucariótico de Iniciación/genética , Factor 4A Eucariótico de Iniciación/metabolismo , Mamíferos/metabolismo , Ratones , Proteínas/metabolismo , ARN/metabolismo , Esteroles/metabolismo , Esteroles/farmacología
2.
Cell Microbiol ; 22(7): e13213, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32353188

RESUMEN

Numerous bacterial toxins exert their activity by inactivating or modulating a specific intracellular host target. For this purpose, these toxins have developed efficient strategies to overcome the different host cell defences including specific binding to cell surface, internalisation, passage through the endosome or plasma membrane, exploiting intracellular trafficking and addressing to intracellular targets. Several intracellularly active toxins deliver an active domain into the cytosol that interacts with a target localised to the inner face of the plasma membrane. Thus, the large clostridial glucosylating toxins (LCGTs) target Rho/Ras-GTPases, certain virulence factors of Gram negative bacteria, Rho-GTPases, while Pasteurella multocida toxin (PMT) targets trimeric G-proteins. Others such as botulinum neurotoxins and tetanus neurotoxin have their substrate on synaptic vesicle membrane. LCGTs, PMT, and certain virulence factors from Vibrio sp. show a particular structure constituted of a four-helix bundle membrane (4HBM) protruding from the catalytic site that specifically binds to the membrane phospholipids and then trap the catalytic domain at the proximity of the membrane anchored substrate. Structural and functional analysis indicate that the 4HBM tip of the Clostridium sordellii lethal toxin (TcsL) from the LCGT family contain two loops forming a cavity that mediates the binding to phospholipids and more specifically to phosphatidylserine.


Asunto(s)
Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacología , Membrana Celular/efectos de los fármacos , Citoplasma/microbiología , Animales , Proteínas Bacterianas , Toxinas Botulínicas , Dominio Catalítico , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Legionella pneumophila , Metaloendopeptidasas , Neurotoxinas , Ácidos Fosfatidicos , Fosfatidilserinas/metabolismo , Toxina Tetánica , Factores de Virulencia/metabolismo , Proteínas de Unión al GTP rho/metabolismo
3.
J Chem Inf Model ; 59(8): 3506-3518, 2019 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-31287306

RESUMEN

We describe here a method to identify potential binding sites in ensembles of protein structures as obtained by molecular dynamics simulations. This is a highly important task in the context of structure-based drug discovery, and many methods exist for the much simpler case of static structures. However, during molecular dynamics, the cavities and grooves that are used to define binding sites merge, split, appear, and disappear, and cover a large volume. Combined with the large number of sites (∼105 and more), these characteristics hamper a consistent and comprehensive definition of binding sites. Our method is based on the calculation of instantaneous cavities and of the pockets delineating them. Classification of the pockets over the structure ensemble generates consensus pockets, which define sites. Sites are reported as lists of atoms or residues. This avoids the pitfalls of the classification of cavities by spatial overlap, used in most existing methods, which is bound to fail on nonordered or unaligned ensembles or as soon as significant molecular motions are involved. To achieve a robust and consistent classification, we thoroughly optimized and benchmarked the method. For this, we assembled from the literature a set of reference sites on systems involving significant functional molecular motions. We tested different descriptors, metrics, and clustering methods. The resulting method is able to perform a global analysis of potential sites efficiently. Tests on examples show that our approach can make predictions of potential sites on the whole surface of a protein and identify novel sites absent from static structures.


Asunto(s)
Modelos Moleculares , Proteínas/química , Proteínas/metabolismo , Sitios de Unión , Conformación Proteica
4.
Bioinformatics ; 31(9): 1490-2, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25543048

RESUMEN

MOTIVATION: Sampling the conformational space of biological macromolecules generates large sets of data with considerable complexity. Data-mining techniques, such as clustering, can extract meaningful information. Among them, the self-organizing maps (SOMs) algorithm has shown great promise; in particular since its computation time rises only linearly with the size of the data set. Whereas SOMs are generally used with few neurons, we investigate here their behavior with large numbers of neurons. RESULTS: We present here a python library implementing the full SOM analysis workflow. Large SOMs can readily be applied on heavy data sets. Coupled with visualization tools they have very interesting properties. Descriptors for each conformation of a trajectory are calculated and mapped onto a 3D landscape, the U-matrix, reporting the distance between neighboring neurons. To delineate clusters, we developed the flooding algorithm, which hierarchically identifies local basins of the U-matrix from the global minimum to the maximum. AVAILABILITY AND IMPLEMENTATION: The python implementation of the SOM library is freely available on github: https://github.com/bougui505/SOM. CONTACT: michael.nilges@pasteur.fr or guillaume.bouvier@pasteur.fr SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Conformación Proteica , Programas Informáticos , Algoritmos , Análisis por Conglomerados , Simulación de Dinámica Molecular
5.
Cell Microbiol ; 17(10): 1477-93, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25882477

RESUMEN

Clostridium sordellii lethal toxin (TcsL) is a potent virulence factor belonging to the large clostridial glucosylating toxin family. TcsL enters target cells via receptor-mediated endocytosis and delivers the N-terminal catalytic domain (TcsL-cat) into the cytosol upon an autoproteolytic process. TcsL-cat inactivates small GTPases including Rac and Ras by glucosylation with uridine-diphosphate (UDP)-glucose as cofactor leading to drastic changes in cytoskeleton and cell viability. TcsL-cat was found to preferentially bind to phosphatidylserine (PS)-containing membranes and to increase the glucosylation of Rac anchored to lipid membrane. We here report binding affinity measurements of TcsL-cat for brain PS-containing membranes by surface plasmon resonance and enzyme-linked immunosorbent assay (ELISA). In addition, TcsL-cat bound to phosphatidic acid (PA) and, to a lesser extent, to other anionic lipids, but not to neutral lipids, sphingolipids or sterol. We further show that the lipid unsaturation status influenced TcsL-cat binding to phospholipids, PS with unsaturated acyl chains and PA with saturated acyl chains being the preferred bindingsubstrates. Phospholipid binding site is localized at the N-terminal four helical bundle structure (1-93 domain). However, TcsL-1-93 bound to a broad range of substrates, whereas TcsL-cat, which is the active domain physiologically delivered into the cytosol, selectively bound to PS and PA. Similar findings were observed with the other large clostridial glucosylating toxins from C. difficile, C. novyi and C. perfringens.


Asunto(s)
Toxinas Bacterianas/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfatidilserinas/metabolismo , Aniones/metabolismo , Sitios de Unión , Dominio Catalítico , Membrana Celular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Unión Proteica , Resonancia por Plasmón de Superficie
6.
BMC Bioinformatics ; 16: 93, 2015 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-25888251

RESUMEN

BACKGROUND: Identifying druggable cavities on a protein surface is a crucial step in structure based drug design. The cavities have to present suitable size and shape, as well as appropriate chemical complementarity with ligands. RESULTS: We present a novel cavity prediction method that analyzes results of virtual screening of specific ligands or fragment libraries by means of Self-Organizing Maps. We demonstrate the method with two thoroughly studied proteins where it successfully identified their active sites (AS) and relevant secondary binding sites (BS). Moreover, known active ligands mapped the AS better than inactive ones. Interestingly, docking a naive fragment library brought even more insight. We then systematically applied the method to the 102 targets from the DUD-E database, where it showed a 90% identification rate of the AS among the first three consensual clusters of the SOM, and in 82% of the cases as the first one. Further analysis by chemical decomposition of the fragments improved BS prediction. Chemical substructures that are representative of the active ligands preferentially mapped in the AS. CONCLUSION: The new approach provides valuable information both on relevant BSs and on chemical features promoting bioactivity.


Asunto(s)
Simulación del Acoplamiento Molecular/métodos , Algoritmos , Sitios de Unión , Diseño de Fármacos , Transcriptasa Inversa del VIH/química , Transcriptasa Inversa del VIH/metabolismo , Ligandos , Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-abl/metabolismo
7.
BMC Genomics ; 15 Suppl 7: S5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25573486

RESUMEN

BACKGROUND: Over the last decades, a vast structural knowledge has been gathered on the HIV-1 protease (PR). Noticeably, most of the studies focused the B-subtype, which has the highest prevalence in developed countries. Accordingly, currently available anti-HIV drugs target this subtype, with considerable benefits for the corresponding patients. RESULTS: Herein, we used molecular dynamics simulations to investigate the role of this polymorphism on the interaction of PR with six of its natural cleavage-sites substrates. CONCLUSIONS: With multiple approaches and analyses we identified structural and dynamical determinants associated with the changes found in the binding affinity of the M36I variant. This mutation influences the flexibility of both PR and its complexed substrate. The observed impact of M36I, suggest that combination with other non-B subtype polymorphisms, could lead to major effects on the interaction with the 12 known cleavage sites, which should impact the virion maturation.


Asunto(s)
Proteasa del VIH/genética , Proteasa del VIH/metabolismo , Polimorfismo Genético , Sitios de Unión/genética , Simulación por Computador , Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/metabolismo , Ligandos , Simulación de Dinámica Molecular , Unión Proteica/genética , Especificidad por Sustrato/genética , Proteínas Virales/química , Proteínas Virales/metabolismo
8.
J Chem Inf Model ; 54(1): 289-301, 2014 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-24397493

RESUMEN

The VanA D-Ala:D-Lac ligase is a key enzyme in the emergence of high level resistance to vancomycin in Enterococcus species and methicillin-resistant Staphylococcus aureus. It catalyzes the formation of D-Ala-D-Lac instead of the vancomycin target, D-Ala-D-Ala, leading to the production of modified, low vancomycin binding affinity peptidoglycan precursors. Therefore, VanA appears as an attractive target for the design of new antibacterials to overcome resistance. The catalytic site of VanA is delimited by three domains and closed by an ω-loop upon enzymatic reaction. The aim of the present work was (i) to investigate the conformational transition of VanA associated with the opening of its ω-loop and of a part of its central domain and (ii) to relate this transition with the substrate or product binding propensities. Molecular dynamics trajectories of the VanA ligase of Enterococcus faecium with or without a disulfide bridge distant from the catalytic site revealed differences in the catalytic site conformations with a slight opening. Conformations were clustered with an original machine learning method, based on self-organizing maps (SOM), which revealed four distinct conformational basins. Several ligands related to substrates, intermediates, or products were docked to SOM representative conformations with the DOCK 6.5 program. Classification of ligand docking poses, also performed with SOM, clearly distinguished ligand functional classes: substrates, reaction intermediates, and product. This result illustrates the acuity of the SOM classification and supports the quality of the DOCK program poses. The protein-ligand interaction features for the different classes of poses will guide the search and design of novel inhibitors.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Ligasas de Carbono-Oxígeno/química , Ligasas de Carbono-Oxígeno/metabolismo , Modelos Moleculares , Inteligencia Artificial , Dominio Catalítico , Biología Computacional , Cristalografía por Rayos X , Diseño de Fármacos , Enterococcus faecium/enzimología , Ligandos , Simulación de Dinámica Molecular , Conformación Proteica , Programas Informáticos , Resistencia a la Vancomicina
9.
Eur J Med Chem ; 269: 116308, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38503166

RESUMEN

Plasmodium multi-resistance, including against artemisinin, seriously threatens malaria treatment and control. Hence, new drugs are urgently needed, ideally targeting different parasitic stages, which are not yet targeted by current drugs. The SUB1 protease is involved in both hepatic and blood stages due to its essential role in the egress of parasites from host cells, and, as potential new target, it would meet the above criteria. We report here the synthesis as well as the biological and structural evaluation of substrate-based α-ketoamide SUB1 pseudopeptidic inhibitors encompassing positions P4-P2'. By individually substituting each position of the reference compound 1 (MAM-117, Ac-Ile-Thr-Ala-AlaCO-Asp-Glu (Oall)-NH2), we better characterized the structural determinants for SUB1 binding. We first identified compound 8 with IC50 values of 50 and 570 nM against Pv- and PfSUB1, respectively (about 3.5-fold higher potency compared to 1). Compound 8 inhibited P. falciparum merozoite egress in culture by 37% at 100 µM. By increasing the overall hydrophobicity of the compounds, we could improve the PfSUB1 inhibition level and antiparasitic activity, as shown with compound 40 (IC50 values of 12 and 10 nM against Pv- and PfSUB1, respectively, IC50 value of 23 µM on P. falciparum merozoite egress). We also found that 8 was highly selective towards SUB1 over three mammalian serine peptidases, supporting the promising value of this compound. Finally, several crystal 3D-structures of SUB1-inhibitor complexes, including with 8, were solved at high resolution to decipher the binding mode of these compounds.


Asunto(s)
Antimaláricos , Malaria Falciparum , Malaria , Parásitos , Animales , Subtilisina/metabolismo , Secuencia de Aminoácidos , Plasmodium falciparum/metabolismo , Péptidos , Malaria Falciparum/parasitología , Serina Proteasas/metabolismo , Relación Estructura-Actividad , Antimaláricos/farmacología , Antimaláricos/química , Proteínas Protozoarias , Mamíferos/metabolismo
10.
Proc Natl Acad Sci U S A ; 107(25): 11277-82, 2010 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-20534570

RESUMEN

Allostery plays a key role in the regulation of the activity and function of many biomolecules. And although many ligands act through allostery, no systematic use is made of it in drug design strategies. Here we describe a procedure for identifying the regions of a protein that can be used to control its activity through allostery. This procedure is based on the construction of a plausible conformational path, which describes protein transition between known active and inactive conformations. The path is calculated by using a framework approach that steers and markedly improves the conjugate peak refinement method. The evolution of conformations along this path was used to identify a putative allosteric site that could regulate activation of Bacillus anthracis adenylyl cyclase toxin (EF) by calmodulin. Conformations of the allosteric site at different steps along the path from the inactive (free) to the active (bound to calmodulin) forms of EF were used to perform virtual screenings and propose candidate EF inhibitors. Several candidates then proved to inhibit calmodulin-induced activation in an in vitro assay. The most potent compound fully inhibited EF at a concentration of 10 microM. The compounds also inhibited the related adenylyl cyclase toxin from Bordetella pertussis (CyaA). The specific homology between the putative allosteric sites in both toxins supports that these pockets are the actual binding sites of the selected inhibitors.


Asunto(s)
Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Calmodulina/química , Sitio Alostérico , Toxinas Bacterianas/antagonistas & inhibidores , Bordetella pertussis/metabolismo , Química Farmacéutica/métodos , Biología Computacional/métodos , Bases de Datos de Proteínas , Diseño de Fármacos , Humanos , Modelos Moleculares , Conformación Proteica , Estructura Terciaria de Proteína
11.
Acta Crystallogr D Struct Biol ; 79(Pt 8): 721-734, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37428845

RESUMEN

The constant selection and propagation of multi-resistant Plasmodium sp. parasites require the identification of new antimalarial candidates involved in as-yet untargeted metabolic pathways. Subtilisin-like protease 1 (SUB1) belongs to a new generation of drug targets because it plays a crucial role during egress of the parasite from infected host cells at different stages of its life cycle. SUB1 is characterized by an unusual pro-region that tightly interacts with its cognate catalytic domain, thus precluding 3D structural analysis of enzyme-inhibitor complexes. In the present study, to overcome this limitation, stringent ionic conditions and controlled proteolysis of recombinant full-length P. vivax SUB1 were used to obtain crystals of an active and stable catalytic domain (PvS1Cat) without a pro-region. High-resolution 3D structures of PvS1Cat, alone and in complex with an α-ketoamide substrate-derived inhibitor (MAM-117), showed that, as expected, the catalytic serine of SUB1 formed a covalent bond with the α-keto group of the inhibitor. A network of hydrogen bonds and hydrophobic interactions stabilized the complex, including at the P1' and P2' positions of the inhibitor, although P' residues are usually less important in defining the substrate specificity of subtilisins. Moreover, when associated with a substrate-derived peptidomimetic inhibitor, the catalytic groove of SUB1 underwent significant structural changes, particularly in its S4 pocket. These findings pave the way for future strategies for the design of optimized SUB1-specific inhibitors that may define a novel class of antimalarial candidates.


Asunto(s)
Antimaláricos , Subtilisina , Plasmodium vivax , Antimaláricos/farmacología , Antimaláricos/química , Inhibidores Enzimáticos/farmacología , Proteínas Protozoarias/química
12.
J Glob Antimicrob Resist ; 28: 84-89, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34929377

RESUMEN

OBJECTIVES: Chagas disease, caused by the parasitic protozoan Trypanosoma cruzi, affects approximately 6-7 million people worldwide. There are limited available therapies and they exhibit low efficacy, often high toxicity in chronic cases and some drug resistance. In this study, our objective was to develop ester prodrugs that inhibit proline racemase (TcPRAC), a parasitic enzyme previously identified and characterised as a promising target because of its essential role in the parasite's life cycle and virulence, and to test their activity against T. cruzi. METHODS: Using structural bioinformatics, we modelled several functional intermediates of the catalytic site between the opened and closed conformations of TcPRAC based on its crystal structures in complex with its competitive inhibitor, pyrrole-2-carboxylic acid. Guided by these intermediates, which were later validated in cocrystals, we designed and evaluated numerous compounds and tested them enzymatically on live parasites and in mice with our quick and straightforward drug screening method, which is based on state-of-the-art bioluminescent T. cruzi parasites injected subcutaneously. RESULTS: Some of our novel compounds specifically inhibited racemase activity, as determined through biochemical assays, and covalently bound to TcPRAC. Furthermore, the corresponding ester prodrugs were effective in killing parasites in vitro. Bioluminescent T. cruzi assays in mice showed that JR1531, a TcPRAC inhibitor prodrug, can kill parasites in living animals, with boosted action when combined with low doses of benznidazole. CONCLUSION: This approach, based on TcPRAC inhibitor prodrugs in association with low doses of benznidazole, may lead to more effective, specific and non-toxic therapies against Chagas disease.


Asunto(s)
Enfermedad de Chagas , Parásitos , Profármacos , Trypanosoma cruzi , Isomerasas de Aminoácido , Animales , Enfermedad de Chagas/tratamiento farmacológico , Ésteres/farmacología , Ésteres/uso terapéutico , Humanos , Ratones , Nitroimidazoles , Profármacos/farmacología , Profármacos/uso terapéutico
13.
Chem Biol Drug Des ; 99(4): 513-526, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34918458

RESUMEN

Proline racemases (PRAC), catalyzing the l-proline and d-proline interconversion, are essential factors in eukaryotic pathogens such as Trypanosoma cruzi, Trypanosoma vivax, and Clostridioides difficile. If the discovery of irreversible inhibitors of T. cruzi PRAC (TcPRAC) led to innovative therapy of the Chagas disease, no inhibitors of CdPRAC have been discovered to date. However, C. difficile, due to an increased incidence in recent years, is considered as a major cause of health threat. In this work, we have taken into account the similarity between TcPRAC and CdPRAC enzymes to design new inhibitors of CdPRAC. Starting from (E) 4-oxopent-2-enoic acid TcPRAC irreversible inhibitors, we synthesized 4-aryl substituted analogs and evaluated their CdPRAC enzymatic inhibition against eleven strains of C. difficile. This study resulted in promising candidates and allowed for identification of (E)-4-(3-bromothiophen-2-yl)-4-oxobut-2-enoic acid 20 that was chosen for complementary in vivo studies and did not reveal in vivo toxicity.


Asunto(s)
Isomerasas de Aminoácido , Antibacterianos , Clostridioides difficile , Isomerasas de Aminoácido/antagonistas & inhibidores , Antibacterianos/farmacología , Clostridioides difficile/efectos de los fármacos , Prolina
14.
Proteins ; 79(5): 1649-61, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21425348

RESUMEN

The anthrax edema factor is a toxin overproducing damaging levels of cyclic adenosine monophosphate (cAMP) and pyrophosphate (PPi) from ATP. Here, mechanisms of dissociation of ATP and products (cAMP, PPi) from the active site are studied using locally enhanced sampling (LES) and steered molecular dynamics simulations. Various substrate conformations and ionic binding modes found in crystallographic structures are considered. LES simulations show that PPi and cAMP dissociate through different solvent accessible channels, while ATP dissociation requires significant active site exposure to solvent. The ionic content of the active site directly affects the dissociation of ATP and products. Only one ion dissociates along with ATP in the two-Mg(2+) binding site, suggesting that the other ion binds EF prior to ATP association. Dissociation of reaction products cAMP and PPi is impaired by direct electrostatic interactions between products and Mg(2+) ions. This provides an explanation for the inhibitory effect of high Mg(2+) concentrations on EF enzymatic activity. Breaking of electrostatic interactions is dependent on a competitive binding of water molecules to the ions, and thus on the solvent accessibility of the active site. Consequently, product dissociation seems to be a two-step process. First, ligands are progressively solvated while preserving the most important electrostatic interactions, in a process that is dependent on the flexibility of the active site. Second, breakage of the electrostatic bonds follows, and ligands diffuse into solvent. In agreement with this mechanism, product protonation facilitates dissociation.


Asunto(s)
Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Adenosina Trifosfato/metabolismo , Antígenos Bacterianos/química , Bacillus anthracis/química , Toxinas Bacterianas/química , AMP Cíclico/metabolismo , Magnesio/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica
15.
Biophys J ; 99(7): 2264-72, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20923661

RESUMEN

Calmodulin (CaM) is a remarkably flexible protein which can bind multiple targets in response to changes in intracellular calcium concentration. It contains four calcium-binding sites, arranged in two globular domains. The calcium affinity of CaM N-terminal domain (N-CaM) is dramatically reduced when the complex with the edema factor (EF) of Bacillus anthracis is formed. Here, an atomic explanation for this reduced affinity is proposed through molecular dynamics simulations and free energy perturbation calculations of the EF-CaM complex starting from different crystallographic models. The simulations show that electrostatic interactions between CaM and EF disfavor the opening of N-CaM domains usually induced by calcium binding. Relative calcium affinities of the N-CaM binding sites are probed by free energy perturbation, and dissociation probabilities are evaluated with locally enhanced sampling simulations. We show that EF impairs calcium binding on N-CaM through a direct conformational restraint on Site 1, by an indirect destabilization of Site 2, and by reducing the cooperativity between the two sites.


Asunto(s)
Antígenos Bacterianos/metabolismo , Bacillus anthracis/química , Toxinas Bacterianas/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Antígenos Bacterianos/química , Toxinas Bacterianas/química , Calmodulina/química , Simulación por Computador , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Termodinámica
16.
Front Mol Biosci ; 7: 586544, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33344505

RESUMEN

Molecular dynamics (MD) simulations have been recorded on the complex between the edema factor (EF) of Bacilllus anthracis and calmodulin (CaM), starting from a structure with the orthosteric inhibitor adefovir bound in the EF catalytic site. The starting structure has been destabilized by alternately suppressing different co-factors, such as adefovir ligand or ions, revealing several long-distance correlations between the conformation of CaM, the geometry of the CaM/EF interface, the enzymatic site and the overall organization of the complex. An allosteric communication between CaM/EF interface and the EF catalytic site, highlighted by these correlations, was confirmed by several bioinformatics approaches from the literature. A network of hydrogen bonds and stacking interactions extending from the helix V of of CaM, and the residues of the switches A, B and C, and connecting to catalytic site residues, is a plausible candidate for the mediation of allosteric communication. The greatest variability in volume between the different MD conditions was also found for cavities present at the EF/CaM interface and in the EF catalytic site. The similarity between the predictions from literature and the volume variability might introduce the volume variability as new descriptor of allostery.

17.
Biophys J ; 96(4): 1249-63, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19217845

RESUMEN

We have studied the relationship between dynamical correlations and energetic contributions in an attempt to model the transmission of information inside protein-protein complexes. The complex formed between the edema factor (EF) of Bacillus anthracis and calmodulin (CaM) was taken as an example, as the formation and stability of the complex depend on the calcium complexation level. The effect of calcium through EF-CaM residue network has been investigated with various approaches: 1), the elastic network model; 2), the local feature analysis; 3), the generalized correlations; and 4), the energetic dependency maps (EDMs), on 15-ns molecular dynamics simulations of the complex loaded with 0, 2, or 4 Ca2+ ions. The elastic network model correctly describes the basic architecture of the complex but is poorly sensitive to the level of calcium compared to the other methods. The local feature analysis allows us to characterize the local dynamics of the complex and the propagation of the calcium signal through CaM. The analyses of global dynamics and energetics--through generalized correlations and EDMs--provide a comprehensive picture of EF-CaM architecture and can be unified by using the concept of residue network connectedness. A medium connectedness, defined as the ability of each residue to communicate with all remaining parts of the complex, is observed for the 2Ca2+ level, which was experimentally identified as the most stable form of EF-CaM. The hierarchy of relative stabilities given by the EDMs sheds a new light on the EF-CaM interaction mechanism described experimentally and supports an organization of the complex architecture centered around nucleation points.


Asunto(s)
Antígenos Bacterianos/química , Toxinas Bacterianas/química , Calcio/química , Calmodulina/química , Análisis por Conglomerados , Simulación por Computador , Cristalografía por Rayos X , Modelos Químicos , Modelos Moleculares , Estructura Cuaternaria de Proteína , Termodinámica
18.
Proteins ; 77(4): 971-83, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19705488

RESUMEN

The Edema Factor (EF), one of the virulence factors of anthrax, is an adenylyl cyclase that promotes the overproduction of cyclic-AMP (cAMP) from ATP, and therefore perturbs cell signaling. Crystallographic structures of EF bound to ATP analogs and reaction products, cyclic-AMP, and Pyrophosphate (PPi), revealed different substrate conformations and catalytic-cation binding modes, one or two cations being observed in the active site. To shed light into the biological significance of these crystallographic structures, the energetics, geometry, and dynamics of the active site are analyzed using molecular dynamics simulations. The ATP conformation observed in the one-metal-ion structure allows stronger interactions with the catalytic ion, and ATP is more restrained than in the structure containing two Mg(2+) ions. Therefore, we propose that the conformation observed in the one-ion crystal structure is a more probable starting point for the reaction. The simulations also suggest that a C3'-endo sugar pucker facilitates nucleophilic attack. Additionally, the two-cation binding mode restrains the mobility of the reaction products, and thus their tendency to dissociate.


Asunto(s)
Adenilil Ciclasas/química , Adenilil Ciclasas/metabolismo , Antígenos Bacterianos/química , Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Magnesio/química , Magnesio/metabolismo , Modelos Moleculares , Conformación Molecular , Complejos Multiproteicos/química , Termodinámica
19.
Proteins ; 71(4): 1813-29, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18175311

RESUMEN

We analyzed the conformational plasticity of calmodulin (CaM) when it is bound to the oedema factor (EF) of Bacillus anthracis and its response to calcium complexation with molecular dynamics (MD) simulations. The EF-CaM complex was simulated during 15 ns for three different levels of calcium bound to CaM. They were respectively no calcium ion (EF-(Apo-CaM)), two calcium ions bound to the C-terminal domain of CaM (EF-(2Ca-CaM)), and four calcium ions bound to CaM (EF-(4Ca-CaM)). Calculations were performed using AMBER package. The analysis of the MD simulations illustrates how CaM forces EF in an open conformation to form the adenylyl cyclase enzymatic site, especially with the two calcium form of CaM, best suited to fit the open conformation of EF. By contrast, CaM encounters bending and unwinding of its flexible interlinker in EF-(Apo-CaM) and EF-(4Ca-CaM). Calcium binding to one domain of CaM affects the other one, showing a transmission of information along the protein structure. The analysis of the CaM domains conformation along the simulations brings an atomistic and dynamic explanation for the instability of these complexes. Indeed the EF-hand helices of the N-terminal domain tend to open upon calcium binding (EF-(4Ca-CaM)), although the domain is locked by EF. By contrast, the C-terminal domain is strongly locked in the open conformation by EF, and the removal of calcium induces a collapse of EF catalytic site (EF-(Apo-CaM)).


Asunto(s)
Antígenos Bacterianos/metabolismo , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Conformación Proteica , Secuencia de Aminoácidos , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Calmodulina/química , Simulación por Computador , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Presión , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Electricidad Estática , Temperatura , Factores de Tiempo , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA