Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Comput Aided Mol Des ; 31(11): 995-1007, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28994029

RESUMEN

Cdc25 phosphatase B, a potential target for cancer therapy, is inhibited by a series of quinones. The binding site and mode of quinone inhibitors to Cdc25B remains unclear, whereas this information is important for structure-based drug design. We investigated the potential binding site of NSC663284 [DA3003-1 or 6-chloro-7-(2-morpholin-4-yl-ethylamino)-quinoline-5, 8-dione] through docking and molecular dynamics simulations. Of the two main binding sites suggested by docking, the molecular dynamics simulations only support one site for stable binding of the inhibitor. Binding sites in and near the Cdc25B catalytic site that have been suggested previously do not lead to stable binding in 50 ns molecular dynamics (MD) simulations. In contrast, a shallow pocket between the C-terminal helix and the catalytic site provides a favourable binding site that shows high stability. Two similar binding modes featuring protein-inhibitor interactions involving Tyr428, Arg482, Thr547 and Ser549 are identified by clustering analysis of all stable MD trajectories. The relatively flexible C-terminal region of Cdc25B contributes to inhibitor binding. The binding mode of NSC663284, identified through MD simulation, likely prevents the binding of protein substrates to Cdc25B. The present results provide useful information for the design of quinone inhibitors and their mechanism of inhibition.


Asunto(s)
Antineoplásicos/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Quinolonas/química , Quinonas/química , Fosfatasas cdc25/antagonistas & inhibidores , Fosfatasas cdc25/química , Sitios de Unión , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
2.
Faraday Discuss ; 169: 477-99, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25340314

RESUMEN

Recent advances in computational hardware, software and algorithms enable simulations of protein-ligand complexes to achieve timescales during which complete ligand binding and unbinding pathways can be observed. While observation of such events can promote understanding of binding and unbinding pathways, it does not alone provide information about the molecular drivers for protein-ligand association, nor guidance on how a ligand could be optimised to better bind to the protein. We have developed the waterswap (C. J. Woods et al., J. Chem. Phys., 2011, 134, 054114) absolute binding free energy method that calculates binding affinities by exchanging the ligand with an equivalent volume of water. A significant advantage of this method is that the binding free energy is calculated using a single reaction coordinate from a single simulation. This has enabled the development of new visualisations of binding affinities based on free energy decompositions to per-residue and per-water molecule components. These provide a clear picture of which protein-ligand interactions are strong, and which active site water molecules are stabilised or destabilised upon binding. Optimisation of the algorithms underlying the decomposition enables near-real-time visualisation, allowing these calculations to be used either to provide interactive feedback to a ligand designer, or to provide run-time analysis of protein-ligand molecular dynamics simulations.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas/química , Agua/química , Algoritmos , Ligandos , Unión Proteica , Termodinámica , Trombina/química
3.
Sci Rep ; 3: 3561, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24356381

RESUMEN

The emergence of a novel H7N9 avian influenza that infects humans is a serious cause for concern. Of the genome sequences of H7N9 neuraminidase available, one contains a substitution of arginine to lysine at position 292, suggesting a potential for reduced drug binding efficacy. We have performed molecular dynamics simulations of oseltamivir, zanamivir and peramivir bound to H7N9, H7N9-R292K, and a structurally related H11N9 neuraminidase. They show that H7N9 neuraminidase is structurally homologous to H11N9, binding the drugs in identical modes. The simulations reveal that the R292K mutation disrupts drug binding in H7N9 in a comparable manner to that observed experimentally for H11N9-R292K. Absolute binding free energy calculations with the WaterSwap method confirm a reduction in binding affinity. This indicates that the efficacy of antiviral drugs against H7N9-R292K will be reduced. Simulations can assist in predicting disruption of binding caused by mutations in neuraminidase, thereby providing a computational 'assay.'


Asunto(s)
Ciclopentanos/farmacología , Farmacorresistencia Viral/genética , Guanidinas/farmacología , Neuraminidasa/genética , Oseltamivir/farmacología , Zanamivir/farmacología , Ácidos Carbocíclicos , Antivirales/farmacología , Biología Computacional , Inhibidores Enzimáticos/farmacología , Humanos , Subtipo H7N9 del Virus de la Influenza A/genética , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Simulación de Dinámica Molecular , Mutación , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/química , Unión Proteica/genética
4.
Biochemistry ; 52(45): 8150-64, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24128064

RESUMEN

The emergence of influenza drug resistance is a major public health concern. The molecular basis of resistance to oseltamivir (Tamiflu) is investigated using a computational assay involving multiple 500 ns unrestrained molecular dynamics (MD) simulations of oseltamivir complexed with mutants of H1N1-2009 influenza neuraminidase. The simulations, accelerated using graphics processors (GPUs), and using a fully explicit model of water, are of sufficient length to observe multiple drug unbinding and rebinding events. Drug unbinding occurs during simulations of known oseltamivir-resistant mutants of neuraminidase. Molecular-level rationalizations of drug resistance are revealed by analysis of these unbinding trajectories, with particular emphasis on the dynamics of the mutant residues. The results indicate that MD simulations can predict weakening of binding associated with drug resistance. In addition, visualization and analysis of binding site water molecules reveal their importance in stabilizing the binding mode of the drug. Drug unbinding is accompanied by conformational changes, driven by the mutant residues, which results in flooding of a key pocket containing tightly bound water molecules. This displaces oseltamivir, allowing the tightly bound water molecules to be released into bulk. In addition to the role of water, analysis of the trajectories reveals novel behavior of the structurally important 150-loop. Motion of the loop, which can move between an open and closed conformation, is intimately associated with drug unbinding and rebinding. Opening of the loop occurs coincidentally with drug unbinding, and interactions between oseltamivir and the loop seem to aid in the repositioning of the drug back into an approximation of its original binding mode on rebinding. The similarity of oseltamivir to a transition state analogue for neuraminidase suggests that the dynamics of the loop could play an important functional role in the enzyme, with loop closing aiding in binding of the substrate and loop opening aiding the release of the product.


Asunto(s)
Antivirales/farmacología , Neuraminidasa/metabolismo , Oseltamivir/farmacología , Sitios de Unión , Farmacorresistencia Viral/genética , Farmacorresistencia Viral/fisiología , Humanos , Simulación de Dinámica Molecular , Mutación , Neuraminidasa/genética , Unión Proteica , Termodinámica
5.
Biochemistry ; 51(21): 4364-75, 2012 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-22574858

RESUMEN

Multidrug resistance of the pandemic H1N1-2009 strain of influenza has been reported due to widespread treatment using the neuraminidase (NA) inhibitors, oseltamivir (Tamiflu), and zanamivir (Relenza). From clinical data, the single I223R (IR(1)) mutant of H1N1-2009 NA reduced efficacy of oseltamivir and zanamivir by 45 and 10 times, (1) respectively. More seriously, the efficacy of these two inhibitors against the double mutant I223R/H275Y (IRHY(2)) was significantly reduced by a factor of 12 374 and 21 times, respectively, compared to the wild-type.(2) This has led to the question of why the efficacy of the NA inhibitors is reduced by the occurrence of these mutations and, specifically, why the efficacy of oseltamivir against the double mutant IRHY was significantly reduced, to the point where oseltamivir has become an ineffective treatment. In this study, 1 µs of molecular dynamics (MD) simulations was performed to answer these questions. The simulations, run using graphical processors (GPUs), were used to investigate the effect of conformational change upon binding of the NA inhibitors oseltamivir and zanamivir in the wild-type and the IR and IRHY mutant strains. These long time scale dynamics simulations demonstrated that the mechanism of resistance of IRHY to oseltamivir was due to the loss of key hydrogen bonds between the inhibitor and residues in the 150-loop. This allowed NA to transition from a closed to an open conformation. Oseltamivir binds weakly with the open conformation of NA due to poor electrostatic interactions between the inhibitor and the active site. The results suggest that the efficacy of oseltamivir is reduced significantly because of conformational changes that lead to the open form of the 150-loop. This suggests that drug resistance could be overcome by increasing hydrogen bond interactions between NA inhibitors and residues in the 150-loop, with the aim of maintaining the closed conformation, or by designing inhibitors that can form a hydrogen bond to the mutant R223 residue, thereby preventing competition between R223 and R152.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H1N1 del Virus de la Influenza A/genética , Neuraminidasa/química , Neuraminidasa/genética , Sustitución de Aminoácidos , Antivirales/farmacología , Gráficos por Computador , Simulación por Computador , Cristalografía por Rayos X , Farmacorresistencia Viral Múltiple/genética , Inhibidores Enzimáticos/farmacología , Historia del Siglo XXI , Humanos , Enlace de Hidrógeno , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Gripe Humana/historia , Gripe Humana/virología , Modelos Moleculares , Simulación de Dinámica Molecular , Neuraminidasa/antagonistas & inhibidores , Oseltamivir/farmacología , Pandemias/historia , Mutación Puntual , Conformación Proteica , Estructura Secundaria de Proteína , Electricidad Estática , Zanamivir/farmacología
6.
J Chem Phys ; 134(5): 054114, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-21303099

RESUMEN

The accurate prediction of absolute protein-ligand binding free energies is one of the grand challenge problems of computational science. Binding free energy measures the strength of binding between a ligand and a protein, and an algorithm that would allow its accurate prediction would be a powerful tool for rational drug design. Here we present the development of a new method that allows for the absolute binding free energy of a protein-ligand complex to be calculated from first principles, using a single simulation. Our method involves the use of a novel reaction coordinate that swaps a ligand bound to a protein with an equivalent volume of bulk water. This water-swap reaction coordinate is built using an identity constraint, which identifies a cluster of water molecules from bulk water that occupies the same volume as the ligand in the protein active site. A dual topology algorithm is then used to swap the ligand from the active site with the identified water cluster from bulk water. The free energy is then calculated using replica exchange thermodynamic integration. This returns the free energy change of simultaneously transferring the ligand to bulk water, as an equivalent volume of bulk water is transferred back to the protein active site. This, directly, is the absolute binding free energy. It should be noted that while this reaction coordinate models the binding process directly, an accurate force field and sufficient sampling are still required to allow for the binding free energy to be predicted correctly. In this paper we present the details and development of this method, and demonstrate how the potential of mean force along the water-swap coordinate can be improved by calibrating the soft-core Coulomb and Lennard-Jones parameters used for the dual topology calculation. The optimal parameters were applied to calculations of protein-ligand binding free energies of a neuraminidase inhibitor (oseltamivir), with these results compared to experiment. These results demonstrate that the water-swap coordinate provides a viable and potentially powerful new route for the prediction of protein-ligand binding free energies.


Asunto(s)
Proteínas/metabolismo , Termodinámica , Agua/metabolismo , Algoritmos , Simulación por Computador , Inhibidores Enzimáticos/farmacología , Virus de la Influenza A/enzimología , Ligandos , Modelos Moleculares , Neuraminidasa/metabolismo , Oseltamivir/farmacología , Unión Proteica
7.
J Mol Graph Model ; 29(3): 347-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21036084

RESUMEN

The outbreaks of chikungunya (CHIKV) and venezuelan equine encephalitis (VEEV) viral infections in humans have emerged or re-emerged in various countries of "Africa and southeast Asia", and "central and south America", respectively. At present, no drug or vaccine is available for the treatment and therapy of both viral infections, but the non-structural protein, nsP3, is a potential target for the design of potent inhibitors that fit at the adenosine-binding site of its macro domain. Here, so as to understand the fundamental basis of the particular interactions between the ADP-ribose bound to the nsP3 amino acid residues at the binding site, molecular dynamics simulations were applied. The results show that these two nsP3 domains share a similar binding pattern for accommodating the ADP-ribose. The ADP-ribose phosphate unit showed the highest degree of stabilization through hydrogen bond interactions with the nsP3 V33 residue and the consequent amino acid residues 110-114. The adenine base of ADP-ribose was specifically recognized by the conserved nsP3 residue D10. Additionally, the ribose and the diphosphate units were found to play more important roles in the CHIKV nsP3-ADP-ribose complex, while the ter-ribose was more important in the VEEV complex. The slightly higher binding affinity of ADP-ribose toward the nsP3 macro domain of VEEV, as predicted by the simulation results, is in good agreement with previous experimental data. These simulation results provide useful information to further assist in drug design and development for these two important viruses.


Asunto(s)
Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Virus Chikungunya/química , Virus de la Encefalitis Equina Venezolana/química , Virus de la Encefalitis Equina Venezolana/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Animales , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Estructura Molecular , Termodinámica , Proteínas no Estructurales Virales/genética
8.
J Chem Inf Model ; 50(8): 1410-7, 2010 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-20726599

RESUMEN

The recent outbreak of the novel 2009 H1N1 influenza in humans has focused global attention on this virus, which could potentially have introduced a more dangerous pandemic of influenza flu. In the initial step of the viral attachment, hemagglutinin (HA), a viral glycoprotein surface, is responsible for the binding to the human SIA alpha2,6-linked sialopentasaccharide host cell receptor (hHAR). Dynamical and structural properties, based on molecular dynamics simulations of the four different HAs of Spanish 1918 (H1-1918), swine 1930 (H1-1930), seasonal 2005 (H1-2005), and a novel 2009 (H1-2009) H1N1 bound to the hHAR were compared. In all four HA-hHAR complexes, major interactions with the receptor binding were gained from HA residue Y95 and the conserved HA residues of the 130-loop, 190-helix, and 220-loop. However, introduction of the charged HA residues K145 and E227 in the 2009 HA binding pocket was found to increase the HA-hHAR binding efficiency in comparison to the three previously recognized H1N1 strains. Changing of the noncharged HA G225 residue to a negatively charged D225 provides a larger number of hydrogen-bonding interactions. The increase in hydrophilicity of the receptor binding region is apparently an evolution of the current pandemic flu from the 1918 Spanish, 1930 swine, and 2005 seasonal strains. Detailed analysis could help the understanding of how different HAs effectively attach and bind with the hHAR.


Asunto(s)
Hemaglutininas Virales/metabolismo , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Oligosacáridos/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Hemaglutininas Virales/química , Interacciones Huésped-Patógeno , Humanos , Enlace de Hidrógeno , Subtipo H1N1 del Virus de la Influenza A/química , Modelos Moleculares , Datos de Secuencia Molecular , Oligosacáridos/química , Infecciones por Orthomyxoviridae/metabolismo , Unión Proteica
9.
Amino Acids ; 39(2): 393-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20037767

RESUMEN

To predict the susceptibility of the probable 2009 influenza A (H1N1-2009) mutant strains to oseltamivir, MD/LIE approach was applied to oseltamivir complexed with the most frequent drug-resistant strains of neuraminidase subtypes N1 and N2: two mutations on the framework residues (N294S and H274Y) and the two others on the direct-binding residues (E119V and R292K) of oseltamivir. Relative to those of the wild type (WT), loss of drug-target interaction energies, especially in terms of electrostatic contributions and hydrogen bonds were dominantly established in the E119V and R292K mutated systems. The inhibitory potencies of oseltamivir towards the WT and mutants were predicted according to the ordering of binding-free energies: WT (-12.3 kcal mol(-1)) > N294S (-10.4 kcal mol(-1)) > H274Y (-9.8 kcal mol(-1)) > E119 V (-9.3 kcal mol(-1)) > R292K (-7.7 kcal mol(-1)), suggesting that the H1N1-2009 influenza with R292K substitution, perhaps, conferred a high level of oseltamivir resistance, while the other mutants revealed moderate resistance levels. This result calls for an urgent need to develop new potent anti-influenza agents against the next pandemic of potentially higher oseltamivir-resistant H1N1-2009 influenza.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/genética , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Oseltamivir/farmacología , Simulación por Computador , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Modelos Moleculares , Mutación , Oseltamivir/metabolismo , Termodinámica
10.
Biophys Chem ; 145(1): 29-36, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19733000

RESUMEN

To reveal the source of oseltamivir-resistance in influenza (A/H5N1) mutants, the drug-target interactions at each functional group were investigated using MD/LIE simulations. Oseltamivir in the H274Y mutation primarily loses the electrostatic and the vdW interaction energies at the -NH(3)(+) and -OCHEt(2) moieties corresponding to the weakened hydrogen-bonds and changed distances to N1 residues. Differentially, the N294S mutation showed small changes of binding energies and intermolecular interactions. Interestingly, the presence of different conformations of E276 positioned between the -OCHEt(2) group and the mutated residue is likely to play an important role in oseltamivir-resistant identification. In the H274Y mutant, it moves towards the -OCHEt(2) group leading to a reduction in hydrophobicity and pocket size, whilst in the N294S mutant it acts as the hydrogen network center bridging with R224 and the mutated residue S294. The molecular details have answered a question of how the H274Y and N294S mutations confer the high- and medium-level of oseltamivir-resistance to H5N1.


Asunto(s)
Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Oseltamivir/uso terapéutico , Virulencia/efectos de los fármacos , Sustitución de Aminoácidos , Antivirales/uso terapéutico , Farmacorresistencia Viral , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Conformación Molecular/efectos de los fármacos , Mutación , Oseltamivir/química , Oseltamivir/farmacología , Virulencia/genética
11.
Biochem Biophys Res Commun ; 385(3): 390-4, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19463784

RESUMEN

The recent outbreak of the novel strain of influenza A (H1N1) virus has raised a global concern of the future risk of a pandemic. To understand at the molecular level how this new H1N1 virus can be inhibited by the current anti-influenza drugs and which of these drugs it is likely to already be resistant to, homology modeling and MD simulations have been applied on the H1N1 neuraminidase complexed with oseltamivir, and the M2-channel with adamantanes bound. The H1N1 virus was predicted to be susceptible to oseltamivir, with all important interactions with the binding residues being well conserved. In contrast, adamantanes are not predicted to be able to inhibit the M2 function and have completely lost their binding with the M2 residues. This is mainly due to the fact that the M2 transmembrane of the new H1N1 strain contains the S31N mutation which is known to confer resistance to adamantanes.


Asunto(s)
Adamantano/análogos & derivados , Antivirales/farmacología , Farmacorresistencia Viral/genética , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/epidemiología , Gripe Humana/virología , Oseltamivir/farmacología , Adamantano/farmacología , Adamantano/uso terapéutico , Antivirales/uso terapéutico , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Mutación , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/genética , Oseltamivir/uso terapéutico , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/genética
12.
J Chem Inf Model ; 49(4): 847-52, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19281265

RESUMEN

Molecular dynamics simulations of the drug-resistant M2 mutants, A30T, S31N, and L26I, were carried out to investigate the inhibition of M2 activity using amantadine (AMT). The closed and open channel conformations were examined via non- and triply protonated H37. For the nonprotonated state, these mutants exhibited zero water density in the conducting region, and AMT was still bound to the channel pore. Thus, water transport is totally suppressed, similar to the wild-type channel. In contrast, the triply protonated states of the mutants exhibited a different water density and AMT position. A30T and L26I both have a greater water density compared to the wild-type M2, while for the A30T system, AMT is no longer inside the pore. Hydrogen bonding between AMT and H37 crucial for the bioactivity is entirely lost in the open conformation. The elimination of this important interaction of these mutations is responsible for the lost of AMT's function in influenza A M2. This is different for the S31N mutant in which AMT was observed to locate at the pore opening region and bond with V27 instead of S31.


Asunto(s)
Amantadina/farmacología , Antivirales/farmacología , Farmacorresistencia Viral/genética , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/genética , Mutación/genética , Simulación por Computador , Canales Iónicos/química , Canales Iónicos/genética , Membrana Dobles de Lípidos , Modelos Moleculares , Permeabilidad , Relación Estructura-Actividad , Agua/química
13.
Proteins ; 76(1): 62-71, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19089976

RESUMEN

Combined quantum mechanical/molecular mechanical (QM/MM) techniques have been applied to investigate the detailed reaction mechanism of the first step of the acylation process by furin in which the cleavage site of the highly pathogenic avian influenza virus subtype H5N1 (HPH5) acts as its substrate. The energy profile shows a simultaneous mechanism, known as a concerted reaction, of the two subprocesses: the proton transfer from Ser368 to His194 and the nucleophilic attack on the carbonyl carbon of the scissile peptide of the HPH5 cleavage site with a formation of tetrahedral intermediate (INT). The calculated energy barrier for this reaction is 16.2 kcal.mol(-1) at QM/MM B3LYP/6-31+G*//PM3-CHARMM22 level of theory. Once the reaction proceeds, the ordering of the electrostatic stabilization by protein environment is of the enzyme-substrate < transition state < INT complexes. Asp153 was found to play the most important role in the enzymatic reaction by providing the highest degree of intermediate complex stabilization. In addition, the negatively charged carbonyl oxygen of INT is well stabilized by the oxyanion hole constructed by Asn295's carboxamide and Ser368's backbone.


Asunto(s)
Furina/metabolismo , Hemaglutininas/metabolismo , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Acilación , Animales , Aves , Simulación por Computador , Cristalografía por Rayos X , Furina/química , Hemaglutininas/química , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Teoría Cuántica , Electricidad Estática , Especificidad por Sustrato
14.
Amino Acids ; 37(4): 725-32, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19002747

RESUMEN

Molecular dynamics simulations were carried out for the mutant oseltamivir-NA complex, to provide detailed information on the oseltamivir-resistance resulting from the H274Y mutation in neuraminidase (NA) of avian influenza H5N1 viruses. In contrast with a previous proposal, the H274Y mutation does not prevent E276 and R224 from forming the hydrophobic pocket for the oseltamivir bulky group. Instead, reduction of the hydrophobicity and size of pocket in the area around an ethyl moiety at this bulky group were found to be the source of the oseltamivir-resistance. These changes were primarily due to the dramatic rotation of the hydrophilic -COO(-) group of E276 toward the ethyl moiety. In addition, hydrogen-bonding interactions with N1 residues at the -NH(3) (+) and -NHAc groups of oseltamivir were replaced by a water molecule. The calculated binding affinity of oseltamivir to NA was significantly reduced from -14.6 kcal mol(-1) in the wild-type to -9.9 kcal mol(-1) in the mutant-type.


Asunto(s)
Antivirales/química , Farmacorresistencia Viral/genética , Inhibidores Enzimáticos/química , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Neuraminidasa/química , Oseltamivir/química , Antivirales/farmacología , Dominio Catalítico , Cristalografía , Inhibidores Enzimáticos/farmacología , Enlace de Hidrógeno , Subtipo H5N1 del Virus de la Influenza A/química , Subtipo H5N1 del Virus de la Influenza A/enzimología , Subtipo H5N1 del Virus de la Influenza A/genética , Simulación de Dinámica Molecular , Mutación/genética , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/genética , Oseltamivir/farmacología , Conformación Proteica
15.
J Theor Biol ; 254(4): 861-7, 2008 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-18706430

RESUMEN

Since the emergence of the severe acute respiratory syndrome (SARS) to date, neither an effective antiviral drug nor a vaccine against SARS is available. However, it was found that a mixture of two HIV-1 proteinase inhibitors, lopinavir and ritonavir, exhibited some signs of effectiveness against the SARS virus. To understand the fine details of the molecular interactions between these proteinase inhibitors and the SARS virus via complexation, molecular dynamics simulations were carried out for the SARS-CoV 3CL(pro) free enzyme (free SARS) and its complexes with lopinavir (SARS-LPV) and ritonavir (SARS-RTV). The results show that flap closing was clearly observed when the inhibitors bind to the active site of SARS-CoV 3CL(pro). The binding affinities of LPV and RTV to SARS-CoV 3CL(pro) do not show any significant difference. In addition, six hydrogen bonds were detected in the SARS-LPV system, while seven hydrogen bonds were found in SARS-RTV complex.


Asunto(s)
Simulación por Computador , Cisteína Endopeptidasas/química , Inhibidores de la Proteasa del VIH/uso terapéutico , Modelos Moleculares , Pirimidinonas/uso terapéutico , Ritonavir/uso terapéutico , Proteínas Virales/química , Catálisis , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/efectos de los fármacos , Enlace de Hidrógeno , Ligandos , Lopinavir , Complejos Multienzimáticos , Unión Proteica , Proteínas Virales/efectos de los fármacos
16.
J Mol Graph Model ; 27(3): 342-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18620883

RESUMEN

To understand how antiviral drugs inhibit the replication of influenza A virus via the M2 ion channel, molecular dynamics simulations have been applied to the six possible protonation states of the M2 ion channel in free form and its complexes with two commercial drugs in a fully hydrated lipid bilayer. Among the six different states of free M2 tetramer, water density was present in the pore of the systems with mono-protonated, di-protonated at adjacent position, tri-protonated and tetra-protonated systems. In the presence of inhibitor, water density in the channel was considerably better reduced by rimantadine than amantadine, agreed well with the experimental IC(50) values. With the preferential position and orientation of the two drugs in all states, two mechanisms of action, where the drug binds to the opening pore and the histidine gate, were clearly explained, i.e., (i) inhibitor was detected to localize slightly closer to the histidine gate and can facilitate the orientation of His37 imidazole rings to lie in the close conformation and (ii) inhibitor acts as a blocker, binding at almost above the opening pore and interacts slightly with the three pore-lining residues, Leu26, Ala30 and Ser31. Here, the inhibitors were found to bind very weakly to the channel due to their allosteric hindrance while theirs side chains were strongly solvated.


Asunto(s)
Amantadina/farmacología , Antivirales/farmacología , Protones , Rimantadina/farmacología , Proteínas de la Matriz Viral/metabolismo , Sitios de Unión , Enlace de Hidrógeno/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Solventes , Agua
17.
Biophys J ; 95(1): 128-34, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18375507

RESUMEN

The origin of the high pathogenicity of an emerging avian influenza H5N1 due to the -RRRKK- insertion at the cleavage loop of the hemagglutinin H5, was studied using the molecular dynamics technique, in comparison with those of the noninserted H5 and H3 bound to the furin (FR) active site. The cleavage loop of the highly pathogenic H5 was found to bind strongly to the FR cavity, serving as a conformation suitable for the proteolytic reaction. With this configuration, the appropriate interatomic distances were found for all three reaction centers of the enzyme-substrate complex: the arrangement of the catalytic triad, attachment of the catalytic Ser(368) to the reactive S1-Arg, and formation of the oxyanion hole. Experimentally, the--RRRKK--insertion was also found to increase in cleavage of hemagglutinin by FR. The simulated data provide a clear answer to the question of why inserted H5 is better cleaved by FR than the other subtypes, explaining the high pathogenicity of avian influenza H5N1.


Asunto(s)
Furina/química , Furina/ultraestructura , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/ultraestructura , Subtipo H5N1 del Virus de la Influenza A/química , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Modelos Químicos , Simulación por Computador , Subtipo H5N1 del Virus de la Influenza A/ultraestructura , Modelos Moleculares , Péptido Hidrolasas/química , Péptido Hidrolasas/ultraestructura
18.
Proteins ; 71(4): 1908-18, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18175324

RESUMEN

To provide detailed information and insight into the drug-target interaction, structure, solvation, and dynamic and thermodynamic properties, the three known-neuraminidase inhibitors-oseltamivir (OTV), zanamivir (ZNV), and peramivir (PRV)-embedded in the catalytic site of neuraminidase (NA) subtype N1 were studied using molecular dynamics simulations. In terms of ligand conformation, there were major differences in the structures of the guanidinium and the bulky groups. The atoms of the guanidinium group of PRV were observed to form many more hydrogen bonds with the surrounded residues and were much less solvated by water molecules, in comparison with the other two inhibitors. Consequently, D151 lying on the 150-loop (residues 147-152) of group-1 neuraminidase (N1, N4, N5, and N8) was considerably shifted to form direct hydrogen bonds with the --OH group of the PRV, which was located rather far from the 150-loop. For the bulky group, direct hydrogen bonds were detected only between the hydrophilic side chain of ZNV and residues R224, E276, and E277 of N1 with rather weak binding, 20-70% occupation. This is not the case for OTV and PRV, in which flexibility and steric effects due to the hydrophobic side chain lead to the rearrangement of the surrounded residues, that is, the negatively charged side chain of E276 was shifted and rotated to form hydrogen bonds with the positively charged moiety of R224. Taking into account all the ligand-enzyme interaction data, the gas phase MM interaction energy of -282.2 kcal/mol as well as the binding free energy (DeltaG(binding)) of -227.4 kcal/mol for the PRV-N1 are significantly lower than those of the other inhibitors. The ordering of DeltaG(binding) of PRV < ZNV < OTV agrees well with the ordering of experimental IC(50) value.


Asunto(s)
Antivirales/farmacología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Ácidos Carbocíclicos , Algoritmos , Antivirales/química , Antivirales/metabolismo , Sitios de Unión , Catálisis , Simulación por Computador , Ciclopentanos/química , Ciclopentanos/metabolismo , Ciclopentanos/farmacología , Bases de Datos de Proteínas , Guanidinas/química , Guanidinas/metabolismo , Guanidinas/farmacología , Enlace de Hidrógeno , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Ligandos , Modelos Moleculares , Conformación Molecular , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/química , Neuraminidasa/metabolismo , Oseltamivir/química , Oseltamivir/metabolismo , Oseltamivir/farmacología , Presión , Unión Proteica , Estructura Secundaria de Proteína , Electricidad Estática , Temperatura , Termodinámica , Factores de Tiempo , Agua/química , Zanamivir/química , Zanamivir/metabolismo , Zanamivir/farmacología
19.
Biophys J ; 92(3): 798-807, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17085491

RESUMEN

Aiming to understand, at the molecular level, why oseltamivir (OTV) cannot be used for inhibition of human influenza neuraminidase subtype N1 as effectively as for subtypes N2 and N9, molecular dynamics simulations were carried out for the three complexes, OTV-N1, OTV-N2, and OTV-N9. The three-dimensional OTV-N2 and OTV-N9 initial structures were represented by the x-ray structures, whereas that of OTV-N1, whose x-ray structure is not yet solved, was built up using the aligned sequence of H5N1 isolated from humans in Thailand with the x-ray structure of the N2-substrate as the template. In comparison to the OTV-N2 and OTV-N9 complexes, dramatic changes were observed in the OTV conformation in the OTV-N1 complex in which two of its bulky side chains, N-acethyl (-NHAc) and 1-ethylproxy group (-OCHEt2), were rotated to adjust the size to fit into the N1 catalytic site. This change leads directly to the rearrangements of the OTV's environment, which are i), distances to its neighbors, W-178 and E-227, are shorter whereas those to residues R-224, E-276, and E-292 are longer; ii), hydrogen bonds to the two nearest neighbors, R-224 and E-276, are still conserved in distance and number as well as percentage occupation; iii), the calculated ligand/enzyme binding free energies of -7.20, -13.44, and -13.29 kcal/mol agree with their inhibitory activities in terms of the experimental IC50 of 36.1-53.2 nM, 1.9-2.7 nM, and 9.5-17.7 nM for the OTV-N1, OTV-N2, and OTV-N9 complexes, respectively; and iv), hydrogen-bond breaking and creation between the OTV and neighborhood residues are accordingly in agreement with the ligand solvation/desolvation taking place in the catalytic site.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/enzimología , Modelos Químicos , Modelos Moleculares , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/ultraestructura , Oseltamivir/química , Antivirales/química , Sitios de Unión , Catálisis , Simulación por Computador , Relación Dosis-Respuesta a Droga , Activación Enzimática , Unión Proteica , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...