Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biophys J ; 120(1): 168-177, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33248127

RESUMEN

Copper(II) is known to bind in the influenza virus His37 cluster in the homotetrameric M2 proton channel and block the proton current needed for uncoating. Copper complexes based on iminodiacetate also block the M2 proton channel and show reduced cytotoxicity and zebrafish-embryo toxicity. In voltage-clamp oocyte studies using the ubiquitous amantadine-insensitive M2 S31N variant, the current block showed fast and slow phases, in contrast to the single phase found for amantadine block of wild-type M2. Here, we evaluate the mechanism of block by copper adamantyl iminodiacitate and copper cyclooctyl iminodiacitate complexes and address whether the complexes can coordinate with one or more of the His37 imidazoles. The current traces were fitted to parametrized master equations. The energetics of binding and the rate constants suggest that the first step is copper complex binding within the channel, and the slow step in the current block is the formation of a Cu-histidine coordination complex. Solution-phase isothermal titration calorimetry and density functional theory (DFT) calculations indicate that imidazole binds to the copper complexes. Structural optimization using DFT reveals that the complexes fit inside the channel and project the Cu(II) toward the His37 cluster, allowing one imidazole to form a coordination complex with Cu(II). Electrophysiology and DFT studies also show that the complexes block the G34E amantadine-resistant mutant despite some crowding in the binding site by the glutamates.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Amantadina/farmacología , Animales , Antivirales/farmacología , Cobre , Farmacorresistencia Viral , Cinética , Proteínas de la Matriz Viral , Pez Cebra
2.
Biophys J ; 119(9): 1811-1820, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33080223

RESUMEN

The ubiquitous mutation from serine (WT) to asparagine at residue 31 (S31N) in the influenza A M2 channel renders it insensitive to amantadine (AMT) and rimantadine (RMT) block, but it is unknown whether the inhibition results from weak binding or incomplete block. Two-electrode voltage clamp (TEVC) of transfected Xenopus oocytes revealed that the M2 S31N channel is essentially fully blocked by AMT at 10 mM, demonstrating that, albeit weak, AMT binding in a channel results in complete block of its proton current. In contrast, RMT achieves only a modest degree of block in the M2 S31N channel at 1 mM, with very little increase in block at 10 mM, indicating that the RMT binding site in the channel saturates with only modest block. From exponential curve fits to families of proton current wash-in and wash-out traces, the association rate constant (k1) is somewhat decreased for both AMT and RMT in the S31N, but the dissociation rate constant (k2) is dramatically increased compared with WT. The potentials of mean force (PMF) from adaptive biasing force (ABF) molecular dynamics simulations predict that rate constants should be exquisitely sensitive to the charge state of the His37 selectivity filter of M2. With one exception out of eight cases, predictions from the simulations with one and three charged side chains bracket the experimental rate constants, as expected for the acidic bath used in the TEVC assay. From simulations, the weak binding can be accounted for by changes in the potentials of mean force, but the partial block by RMT remains unexplained.


Asunto(s)
Gripe Humana , Rimantadina , Amantadina/farmacología , Antivirales/farmacología , Trastornos Disociativos , Humanos , Proteínas de la Matriz Viral/genética
3.
ACS Chem Biol ; 15(9): 2331-2337, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32786258

RESUMEN

We report on using the synthetic aminoadamantane-CH2-aryl derivatives 1-6 as sensitive probes for blocking M2 S31N and influenza A virus (IAV) M2 wild-type (WT) channels as well as virus replication in cell culture. The binding kinetics measured using electrophysiology (EP) for M2 S31N channel are very dependent on the length between the adamantane moiety and the first ring of the aryl headgroup realized in 2 and 3 and the girth and length of the adamantane adduct realized in 4 and 5. Study of 1-6 shows that, according to molecular dynamics (MD) simulations and molecular mechanics Poisson-Boltzmann surface area (MM/PBSA) calculations, all bind in the M2 S31N channel with the adamantyl group positioned between V27 and G34 and the aryl group projecting out of the channel with the phenyl (or isoxazole in 6) embedded in the V27 cluster. In this outward binding configuration, an elongation of the ligand by only one methylene in rimantadine 2 or using diamantane or triamantane instead of adamantane in 4 and 5, respectively, causes incomplete entry and facilitates exit, abolishing effective block compared to the amantadine derivatives 1 and 6. In the active M2 S31N blockers 1 and 6, the phenyl and isoxazolyl head groups achieve a deeper binding position and high kon/low koff and high kon/high koff rate constants, compared to inactive 2-5, which have much lower kon and higher koff. Compounds 1-5 block the M2 WT channel by binding in the longer area from V27-H37, in the inward orientation, with high kon and low koff rate constants. Infection of cell cultures by influenza virus containing M2 WT or M2 S31N is inhibited by 1-5 or 1-4 and 6, respectively. While 1 and 6 block infection through the M2 block mechanism in the S31N variant, 2-4 may block M2 S31N virus replication in cell culture through the lysosomotropic effect, just as chloroquine is thought to inhibit SARS-CoV-2 infection.


Asunto(s)
Adamantano/farmacología , Virus de la Influenza A/efectos de los fármacos , Gripe Humana/prevención & control , Canales Iónicos/antagonistas & inhibidores , Sondas Moleculares/química , Proteínas de la Matriz Viral/antagonistas & inhibidores , Adamantano/análogos & derivados , Adamantano/química , Adamantano/metabolismo , Betacoronavirus/efectos de los fármacos , Sitios de Unión , COVID-19 , Células Cultivadas , Cloroquina/farmacología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Variación Genética , Humanos , Virus de la Influenza A/química , Virus de la Influenza A/genética , Gripe Humana/tratamiento farmacológico , Cinética , Sondas Moleculares/metabolismo , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Unión Proteica , SARS-CoV-2 , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
4.
Biochim Biophys Acta Biomembr ; 1862(3): 183156, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31846647

RESUMEN

We have investigated the perturbation of influenza A M2TM in DMPC bilayers. We have shown that (a) DSC and SAXS detect changes in membrane organization caused by small changes (micromolar) in M2TM or aminoadamantane concentration and aminoadamantane structure, by comparison of amantadine and spiro[pyrrolidine-2,2'-adamantane] (AK13), (b) that WAXS and MD can suggest details of ligand topology. DSC and SAXS show that at a low M2TM micromolar concentration in DPMC bilayers, two lipid domains are observed, which likely correspond to M2TM boundary lipids and bulk-like lipids. At higher M2TM concentrations, one domain only is identified, which constitutes essentially all of the lipid molecules behaving as boundary lipids. According to SAXS, WAXS, and DSC in the absence of M2TM, both aminoadamantane drugs exert a similar perturbing effect on the bilayer at low concentrations. At the same concentrations of the drug when M2TM is present, amantadine and, to a lesser extent, AK13 cause, according to WAXS, a significant disordering of chain-stacking, which also leads to the formation of two lipid domains. This effect is likely due, according to MD simulations, to the preference of the more lipophilic AK13 to locate closer to the lateral surfaces of M2TM when compared to amantadine, which forms stronger ionic interactions with phosphate groups. The preference of AK13 to concentrate inside the lipid bilayer close to the exterior of the hydrophobic M2TM helices may contribute to its higher binding affinity compared to amantadine.


Asunto(s)
Dimiristoilfosfatidilcolina/química , Membrana Dobles de Lípidos/química , Proteínas de la Matriz Viral/metabolismo , Amantadina/química , Amantadina/farmacología , Antivirales/farmacología , Sitios de Unión , Humanos , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Ligandos , Simulación de Dinámica Molecular , Dominios Proteicos , Dispersión del Ángulo Pequeño , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/fisiología , Difracción de Rayos X
5.
Antiviral Res ; 164: 81-90, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30742842

RESUMEN

In search of novel targets for influenza inhibitors, a site on PB1 was selected for its high conservation and probable interaction with a host protein, RanBP5, that is key to nuclear import of PB1, where it complexes with PB2, PA, and NP to transcribe viral RNA. Docking with libraries of drug-like compounds led to a selection of five candidates that bound tightly and with a pose likely to inhibit protein binding. These were purchased and tested in vitro, found to be active, and then one was synthetically expanded to explore the structure-activity relationship. The top candidates had a carboxylic acid converted to an ester and electron-withdrawing substituents added to a phenyl group in the original structure. Resistance was slow to develop, but cytotoxicity was moderately high. Nuclear localization of PB1 and in vitro polymerase activity were both strongly inhibited.


Asunto(s)
Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Proteínas Virales/antagonistas & inhibidores , beta Carioferinas/metabolismo , Simulación por Computador , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Humanos , Virus de la Influenza A/enzimología , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
6.
ACS Med Chem Lett ; 9(3): 198-203, 2018 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-29541360

RESUMEN

Recently, the binding kinetics of a ligand-target interaction, such as the residence time of a small molecule on its protein target, are seen as increasingly important for drug efficacy. Here, we investigate these concepts to explain binding and proton blockage of rimantadine variants bearing progressively larger alkyl groups to influenza A virus M2 wild type (WT) and M2 S31N protein proton channel. We showed that resistance of M2 S31N to rimantadine analogues compared to M2 WT resulted from their higher koff rates compared to the kon rates according to electrophysiology (EP) measurements. This is due to the fact that, in M2 S31N, the loss of the V27 pocket for the adamantyl cage resulted in low residence time inside the M2 pore. Both rimantadine enantiomers have similar channel blockage and binding kon and koff against M2 WT. To compare the potency between the rimantadine variants against M2, we applied approaches using different mimicry of M2, i.e., isothermal titration calorimetry and molecular dynamics simulation, EP, and antiviral assays. It was also shown that a small change in an amino acid at site 28 of M2 WT, which does not line the pore, seriously affects M2 WT blockage kinetics.

7.
Antiviral Res ; 147: 100-106, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29032206

RESUMEN

New M2 blockers effective against the ubiquitous amantadine-resistant S31N M2 mutation in influenza A are needed. Six copper complexes, 2, 4, 6, 8, 9, and 10, were synthesized and found to block both wild type and S31N M2. Free Cu2+ also blocks M2 S31N but not S31N/H37A. The copper complexes do not block M2 H37A (either S31 or S31N). The complexes were effective against three influenza A strains in cell-culture assays, but less toxic to cells than CuCl2. For example 4, Cu(cyclooctylamineiminodiacetate), which was stable at pH > 4 in the buffers used, had an EC50 against A/Calif/07/2009 H1N1 of 0.7 ± 0.1 µM with a CC50 of 147 µM (therapeutic index, averaged over three strains, 67.8). In contrast, CuCl2 had an EC50 of 3.8 ± 0.9 µM and CC50 of 19 µM. Because M2 H37 is highly conserved, these complexes show promise for further testing as drugs against all strains of influenza A.


Asunto(s)
Antivirales/farmacología , Cobre/farmacología , Farmacorresistencia Viral/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Proteínas de la Matriz Viral/antagonistas & inhibidores , Amantadina/farmacología , Animales , Antivirales/química , Supervivencia Celular/efectos de los fármacos , Cobre/química , Cobre/toxicidad , Perros , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral/genética , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Subtipo H1N1 del Virus de la Influenza A/genética , Dosificación Letal Mediana , Células de Riñón Canino Madin Darby , Mutación , Relación Estructura-Actividad , Índice Terapéutico , Proteínas de la Matriz Viral/genética
8.
J Phys Chem B ; 119(35): 11548-59, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26268449

RESUMEN

The mechanism of amantadine binding to the S31 variant of the M2 protein of Influenza A is well understood, but the reasons behind N31 M2 amantadine insensitivity remain under investigation. Many molecular dynamics studies have evaluated the influence of amantadine position within the channel pore on its ability to inhibit proton conductance in M2, but little is known about the influence of amantadine rotational orientation. Replica-exchange umbrella sampling, steered, and classic molecular dynamics simulations were performed on amantadine in the solid-state NMR structure of S31 M2 and an N31 M2 homologue, both in the homotetramer configuration, to explore the effects of the position and tilt angle of amantadine on inhibition of the M2 channel. Steered simulations show that amantadine rotates with the amine toward the bulk water as it passes into the hydrophobic entryway lined by Val27 side chains. Results from all simulation types performed indicate that amantadine has a strong, specific orientation with the amine turned inward toward the central cavity in the S31 M2 pore but has variable orientation and a strong propensity to remain outward pointing in N31 M2. Free energy profiles from umbrella sampling, measured relative to bulk water, show amantadine binds more strongly to the S31 M2 pore by 8 kcal/mol in comparison to amantadine in the N31 pore, suggesting that it can escape more readily from the N31 channel through the Val27 secondary gate, whereas it is captured by the S31 channel in the same region. Lower water density and distribution near amantadine in S31 M2 reveal that the drug inhibits proton conductance in S31 M2 because of its inward-pointing configuration, whereas in N31 M2, amantadine forms hydrogen bonds with an N31 side chain and does not widely occlude water occupancy in any configuration. Both amantadine's weaker binding to and weaker water occlusion in N31 M2 might contribute to its inefficacy as an inhibitor of the mutant protein.


Asunto(s)
Amantadina/química , Antivirales/química , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética , Agua/química , Amantadina/farmacología , Antivirales/farmacología , Farmacorresistencia Viral/genética , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Virus de la Influenza A , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Rotación
9.
BMC Genet ; 16 Suppl 2: S3, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25953496

RESUMEN

BACKGROUND: The S31N amantadine-resistance mutation in the influenza A M2 sequence currently occurs more frequently in nature than the S31 wild type. Overcoming the resistance of the S31N mutation is the primary focus of M2 researchers who aim to develop novel antiviral therapies. Recent studies have noted a possible rise in frequency of the V27A/S31N double amantadine-resistance mutation in recent years. The purpose of this study is to investigate this recent rise in frequency of the double mutation and any possible bias of the other mutations toward co-occurrence with S31N or S31 strains. RESULTS: The primary dataset used for this study was comprised of 24,152 influenza A M2 channel sequences which were downloaded from UniProt. There is an increased frequency for the S31N/V27A dual AR mutation in recent years, especially in swine. A test for difference in two proportions indicates that the V27A mutation is co-occurring with S31N more often than expected (p-value<0.001) when considering individual amino acid frequencies. At the same time, the different propensities for the V27A as compared to the V27T dual mutant may reflect differences in viral fitness or protein energetics, and this information could be exploited to focus drug development so as to reduce further drug insensitivity. CONCLUSIONS: The development of the S31N/V27A variant in the Midwestern US swine may be a harbinger of novel human strain development. V27A/S31N is a possible path forward for the evolution of M2 which may convey a new level of drug resistance and should receive attention in drug design.


Asunto(s)
Amantadina/farmacología , Farmacorresistencia Viral , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/genética , Mutación Puntual , Proteínas de la Matriz Viral/genética , Animales , Virus de la Influenza A/clasificación , Medio Oeste de Estados Unidos , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Porcinos , Enfermedades de los Porcinos/virología
10.
J Phys Chem B ; 119(3): 1225-31, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25426702

RESUMEN

The mechanisms responsible for drug resistance in the Asn31 variant of the M2 protein of influenza A are not well understood. Molecular dynamics simulations were performed on wild-type (Ser31) and S31N influenza A M2 in the homotetramer configuration. After evaluation of 13 published M2 structures, a solid-state NMR structure with amantadine bound was selected for simulations, an S31N mutant structure was developed and equilibrated, and the native and mutant structures were used to determine the binding behavior of amantadine and the dynamics of water in the two channels. Amantadine is stable in the plugging region of wild-type M2, with the adamantane in contact with the Val27 side chains, while amantadine in S31N M2 has more variable movement and orientation, and spontaneously moves lower into the central cavity of the channel. Free energy profiles from umbrella sampling support this observation. In this configuration, water surrounds the drug and can easily transport protons past it, so the drug binds without blocking proton transport in the S31N M2 channel.


Asunto(s)
Amantadina/metabolismo , Amantadina/farmacología , Farmacorresistencia Viral , Virus de la Influenza A/efectos de los fármacos , Protones , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/metabolismo , Transporte Biológico/efectos de los fármacos , Modelos Moleculares , Conformación Proteica , Proteínas de la Matriz Viral/química , Agua/metabolismo
11.
J Med Chem ; 57(11): 4629-39, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24793875

RESUMEN

A series of 2-adamantanamines with alkyl adducts of various lengths were examined for efficacy against strains of influenza A including those having an S31N mutation in M2 proton channel that confer resistance to amantadine and rimantadine. The addition of as little as one CH2 group to the methyl adduct of the amantadine/rimantadine analogue, 2-methyl-2-aminoadamantane, led to activity in vitro against two M2 S31N viruses A/Calif/07/2009 (H1N1) and A/PR/8/34 (H1N1) but not to a third A/WS/33 (H1N1). Solid state NMR of the transmembrane domain (TMD) with a site mutation corresponding to S31N shows evidence of drug binding. But electrophysiology using the full length S31N M2 protein in HEK cells showed no blockade. A wild type strain, A/Hong Kong/1/68 (H3N2) developed resistance to representative drugs within one passage with mutations in M2 TMD, but A/Calif/07/2009 S31N was slow (>8 passages) to develop resistance in vitro, and the resistant virus had no mutations in M2 TMD. The results indicate that 2-alkyl-2-aminoadamantane derivatives with sufficient adducts can persistently block p2009 influenza A in vitro through an alternative mechanism. The observations of an HA1 mutation, N160D, near the sialic acid binding site in both 6-resistant A/Calif/07/2009(H1N1) and the broadly resistant A/WS/33(H1N1) and of an HA1 mutation, I325S, in the 6-resistant virus at a cell-culture stable site suggest that the drugs tested here may block infection by direct binding near these critical sites for virus entry to the host cell.


Asunto(s)
Adamantano/análogos & derivados , Adamantano/síntesis química , Antivirales/síntesis química , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Adamantano/farmacología , Amantadina/farmacología , Animales , Antivirales/farmacología , Perros , Farmacorresistencia Viral Múltiple , Células HEK293 , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Canales Iónicos/genética , Células de Riñón Canino Madin Darby , Mutación , Rimantadina/farmacología , Proteínas de la Matriz Viral/genética
13.
Curr Opin Virol ; 2(2): 128-33, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22482709

RESUMEN

The M2 protein from influenza A is a proton channel as a tetramer, with a single transmembrane helix from each monomer lining the pore. Val27 and Trp41 form gates at either end of the pore and His37 mediates the shuttling of protons across a central barrier between the N-terminal and C-terminal aqueous pore regions. Numerous structures of this transmembrane domain and of a longer construct that includes an amphipathic helix are now in the Protein Data Bank. Many structural differences are apparent from samples obtained in a variety of membrane mimetic environments. High-resolution structural results in lipid bilayers have provided novel insights into the functional mechanism of the unique HxxxW cluster in the M2 proton channel.


Asunto(s)
Virus de la Influenza A/química , Virus de la Influenza A/metabolismo , Gripe Humana/virología , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo , Biofisica , Humanos , Virus de la Influenza A/genética , Proteínas de la Matriz Viral/genética
14.
BMC Biophys ; 5: 5, 2012 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-22444827

RESUMEN

BACKGROUND: The mechanism of action of volatile general anesthetics has not yet been resolved. In order to identify the effects of isoflurane on the membrane, we measured the steady-state anisotropy of two fluorescent probes that reside at different depths. Incorporation of anesthetic was confirmed by shifting of the main phase transition temperature. RESULTS: In liquid crystalline dipalmitoylphosphatidylcholine liposomes, isoflurane (7-25 mM in the bath) increases trimethylammonium-diphenylhexatriene fluorescence anisotropy by ~0.02 units and decreases diphenylhexatriene anisotropy by the same amount. CONCLUSIONS: The anisotropy data suggest that isoflurane decreases non-axial dye mobility in the headgroup region, while increasing it in the tail region. We propose that these results reflect changes in the lateral pressure profile of the membrane.

15.
Protein J ; 30(7): 490-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21927900

RESUMEN

The protein dipole moment is a low-resolution parameter that characterizes the second-order charge organization of a biomolecule. Theoretical approaches to calculate protein dipole moments rely on pK(a) values, which are either computed individually for each ionizable residue or obtained from model compounds. The influence of pK(a) shifts are evaluated first by comparing calculated and measured dipole moments of ß-lactoglobulin. Second, calculations are made on a dataset of 66 proteins from the Protein Data Bank, and average differences are determined between dipole moments calculated with model pK(a)s, pK(a)s derived using a Poisson-Boltzmann approach, and empirically-calculated pK(a)s. Dipole moment predictions that neglect pK(a) shifts are consistently larger than predictions in which they are included. The importance of pK(a) shifts are observed to vary with protein size, internal permittivity, and solution pH.


Asunto(s)
Lactoglobulinas/química , Concentración de Iones de Hidrógeno , Cinética , Termodinámica
17.
J Phys Chem B ; 115(10): 2205-13, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21344910

RESUMEN

Protein charge organization is dependent on the low-permittivity region in the hydrophobic core of the molecule. We suggest a novel approach to estimate the dielectric constant of this region by comparing measured and simulated first- and second-order charge moments. Here, the dipole moment is measured as a function of pH using dielectric spectroscopy. The results are compared to dipole moments based on Poisson-Boltzmann estimates of pK(a) shifts calculated from structures in the Protein Data Bank. Structures are additionally refined using CHARMM molecular dynamics simulations. The best estimate for the internal permittivity is found by minimizing the root-mean-square residual between measured and predicted charge moments. Using the protein ß-lactoglobulin, a core dielectric constant in the range of 6-7 is estimated.


Asunto(s)
Espectroscopía Dieléctrica/métodos , Electricidad , Proteínas , Ácido Clorhídrico/química , Concentración de Iones de Hidrógeno , Lactoglobulinas/química , Proteínas/química
18.
Biochim Biophys Acta ; 1808(2): 516-21, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20969830

RESUMEN

Amantadine-sensitive proton uptake by liposomes is currently the preferred method of demonstrating M2 functionality after reconstitution, to validate structural determination with techniques such as solid-state NMR. With strong driving forces (two decades each of both [K(+)] gradient-induced membrane potential and [H(+)] gradient), M2(22-62) showed a transport rate of 78 H(+)/tetramer-s (pH(o) 6.0, pH(i) 8.0, nominal V(m)=-114 mV), higher than previously measured for similar, shorter, and full-length constructs. Amantadine sensitivity of the conductance domain at pH 6.8 was also comparable to other published reports. Proton flux rate was optimal at protein densities of 0.05-1.0% (peptide wt.% in lipid). Rundown of total proton uptake after addition of valinomycin and CCCP, as detected by delayed addition of valinomycin, indicated M2-induced K(+) flux of 0.1K(+)/tetramer-s, and also demonstrated that the K(+) permeability, relative to H(+), was 2.8 × 10(-6). Transport rate, amantadine and cyclooctylamine sensitivity, acid activation, and H(+) selectivity were all consistent with full functionality of the reconstituted conductance domain. Decreased external pH increased proton uptake with an apparent pK(a) of 6.


Asunto(s)
Virus de la Influenza A/química , Canales Iónicos/química , Proteínas de la Matriz Viral/química , Amantadina/farmacología , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Concentración de Iones de Hidrógeno , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Cinética , Liposomas , Potenciales de la Membrana , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Valinomicina/farmacología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
19.
Biochim Biophys Acta ; 1808(2): 538-46, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20655872

RESUMEN

Recent controversies associated with the structure of the M2 protein from influenza A virus and the binding site of drug molecules amantadine and rimantadine motivated the comparison here of the drug binding to three viral porins including the M2 proteins from influenza A and B as well as the viral protein 'u' from HIV-1. While the M2 protein from influenza B does not normally bind amantadine, chimeras with the M2 protein from influenza A show blockage by amantadine. Similarly, Vpu does not normally bind rimantadine, but the single site mutation A18H converts a non-specific channel to a selective proton channel that is sensitive to rimantadine. The comparison of structures and amino acid sequences shows that the membrane protein sample environment can have a significant influence on the structural result. While a bilayer surface bound amphipathic helix has been characterized for AM2, such a helix may be possible for BM2 although it has evaded structural characterization in detergent micelles. A similar amphipathic helix seems less likely for Vpu. Even though the A18H Vpu mutant forms rimantadine sensitive proton channels, the binding of drug and its influence on the protein structure appears to be very different from that for the M2 proteins. Indeed, drug binding and drug resistance in these viral porins appears to result from a complex set of factors.


Asunto(s)
Porinas/química , Porinas/genética , Proteínas Virales/química , Proteínas Virales/genética , Secuencia de Aminoácidos , Sitios de Unión , Farmacorresistencia Viral/genética , Conductividad Eléctrica , Genes Virales , VIH-1/química , VIH-1/efectos de los fármacos , VIH-1/genética , Proteínas del Virus de la Inmunodeficiencia Humana/química , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Virus de la Influenza A/química , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/genética , Virus de la Influenza B/química , Virus de la Influenza B/efectos de los fármacos , Virus de la Influenza B/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Homología de Secuencia de Aminoácido , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/genética
20.
Science ; 330(6003): 509-12, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20966252

RESUMEN

The M2 protein from the influenza A virus, an acid-activated proton-selective channel, has been the subject of numerous conductance, structural, and computational studies. However, little is known at the atomic level about the heart of the functional mechanism for this tetrameric protein, a His(37)-Trp(41) cluster. We report the structure of the M2 conductance domain (residues 22 to 62) in a lipid bilayer, which displays the defining features of the native protein that have not been attainable from structures solubilized by detergents. We propose that the tetrameric His(37)-Trp(41) cluster guides protons through the channel by forming and breaking hydrogen bonds between adjacent pairs of histidines and through specific interactions of the histidines with the tryptophan gate. This mechanism explains the main observations on M2 proton conductance.


Asunto(s)
Virus de la Influenza A/química , Canales Iónicos/química , Protones , Proteínas de la Matriz Viral/química , Histidina/química , Enlace de Hidrógeno , Concentración de Iones de Hidrógeno , Virus de la Influenza A/fisiología , Transporte Iónico , Membrana Dobles de Lípidos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Estructura Terciaria de Proteína , Triptófano/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...