Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Comput Aided Mol Des ; 35(1): 95-104, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33037548

RESUMEN

We investigate the binding of native ß-cyclodextrin (ß-CD) and eight novel ß-CD derivatives with two different guest compounds, using isothermal calorimetry and 2D NOESY NMR. In all cases, the stoichiometry is 1:1 and binding is exothermic. Overall, modifications at the 3' position of ß-CD, which is at the secondary face, weaken binding by several kJ/mol relative to native ß-CD, while modifications at the 6' position (primary face) maintain or somewhat reduce the binding affinity. The variations in binding enthalpy are larger than the variations in binding free energy, so entropy-enthalpy compensation is observed. Characterization of the bound conformations with NOESY NMR shows that the polar groups of the guests may be situated at either face, depending on the host molecule, and, in some cases, both orientations are populated. The present results were used in the SAMPL7 blinded prediction challenge whose results are detailed in the same special issue of JCAMD.


Asunto(s)
Ciclodextrinas/metabolismo , Ciclohexanoles/metabolismo , Rimantadina/metabolismo , Termodinámica , beta-Ciclodextrinas/metabolismo , Ciclodextrinas/química , Ciclohexanoles/química , Entropía , Estructura Molecular , Rimantadina/química , beta-Ciclodextrinas/química
2.
Molecules ; 25(17)2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32883012

RESUMEN

A series of nineteen amino acid analogues of amantadine (Amt) and rimantadine (Rim) were synthesized and their antiviral activity was evaluated against influenza virus A (H3N2). Among these analogues, the conjugation of rimantadine with glycine illustrated high antiviral activity combined with low cytotoxicity. Moreover, this compound presented a profoundly high stability after in vitro incubation in human plasma for 24 h. Its thermal stability was established using differential and gravimetric thermal analysis. The crystal structure of glycyl-rimantadine revealed that it crystallizes in the orthorhombic Pbca space group. The structure-activity relationship for this class of compounds was established, with CoMFA (Comparative Molecular Field Analysis) 3D-Quantitative Structure Activity Relationships (3D-QSAR) studies predicting the activities of synthetic molecules. In addition, molecular docking studies were conducted, revealing the structural requirements for the activity of the synthetic molecules.


Asunto(s)
Adamantano/análogos & derivados , Adamantano/farmacología , Antivirales/farmacología , Simulación por Computador , Orthomyxoviridae/efectos de los fármacos , Relación Estructura-Actividad Cuantitativa , Adamantano/síntesis química , Adamantano/química , Animales , Antivirales/síntesis química , Antivirales/química , Sitios de Unión , Muerte Celular/efectos de los fármacos , Cristalografía por Rayos X , Análisis Diferencial Térmico , Perros , Estabilidad de Medicamentos , Humanos , Enlace de Hidrógeno , Análisis de los Mínimos Cuadrados , Células de Riñón Canino Madin Darby , Conformación Molecular , Simulación del Acoplamiento Molecular , Dominios Proteicos , Rimantadina/sangre , Rimantadina/química , Temperatura , Proteínas de la Matriz Viral/química
3.
Molecules ; 24(21)2019 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-31684129

RESUMEN

A zone-fluidics (ZF) based automated fluorimetric sensor for the determination of pharmaceutically active adamantine derivatives, i.e., amantadine (AMA), memantine (MEM) and rimantadine (RIM) is reported. Discrete zones of the analytes and reagents (o-phthalaldehyde and N-acetylcysteine) mix and react under stopped-flow conditions to yield fluorescent iso-indole derivatives (λex/ λem = 340/455 nm). The proposed ZF sensor was developed and validated to prove suitable for quality control tests (assay and content uniformity) of commercially available formulations purchased from the Greek market (EU licensed) and from non-EU web-pharmacies at a sampling rate of 16 h-1. Interestingly, a formulation obtained through the internet and produced in a third-non-EU-country (AMA capsules, 100 mg per cap), was found to be out of specifications (mean assay of 85.3%); a validated HPLC method was also applied for confirmatory purposes.


Asunto(s)
Amantadina/aislamiento & purificación , Fluorometría/métodos , Memantina/aislamiento & purificación , Rimantadina/aislamiento & purificación , Amantadina/química , Cromatografía Líquida de Alta Presión , Indicadores y Reactivos/química , Indoles/química , Memantina/química , Microfluídica , Rimantadina/química
4.
Molecules ; 24(9)2019 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-31083636

RESUMEN

This work discloses a new procedure for the resolution of commercially available racemic rimantadine hydrochloride to enantiomerically pure (S)-rimantadine using (R)-phenoxypropionic acid as a recyclable resolving reagent. Good chemical yields, operational ease, and low-cost structure underscore the preparative value of this method for the production of enantiomerically pure rimantadine for medicinal or synthetic studies.


Asunto(s)
Rimantadina/química , Aminas/química , Estructura Molecular , Estereoisomerismo
5.
Interdiscip Sci ; 11(2): 292-299, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30194627

RESUMEN

HCV p7 protein is a cation-selective ion channel, playing an essential role during the life cycle of HCV viruses. To understand the cation-selective mechanism, we constructed a hexameric model in lipid bilayers of HCV p7 protein for HCB JFH-1 strain, genotype 2a. In this structural model, His9 and Val6 were key factors for the HCV cation-selective ion channel. The histidine residues at position 9 in the hexameric model formed a first gate for HCV p7 channel, acting as a selectivity filter for cations. The valines mentioned above formed a second gate for HCV p7 channel, serving as a hydrophobic filter for the dehydrated cations. The binding pocket for the channel blockers, e.g., amantadine and rimantadine, was composed of residues 20-26 in H2 helix and 52-60 in H3 helix in i + 2 monomer. However, the molecular volumes for both amantadine and rimantadine were too small for the binding pocket of HCV p7 channel. Thus, designing a compound similar with rimantadine and having much larger volume would be an effective strategy for discovering inhibitors against HCV p7 channel. To achieve this point, we used rimantadine as a structural template to search ChEMBL database for the candidates employing favorable binding affinities to HCV p7 channel. As a result, six candidates were identified to have potential to be novel inhibitors against HCV p7 channel.


Asunto(s)
Antivirales/farmacología , Biología Computacional/métodos , Diseño de Fármacos , Hepacivirus/metabolismo , Proteínas Virales/metabolismo , Sitios de Unión , Hepacivirus/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Canales Iónicos/química , Canales Iónicos/metabolismo , Modelos Moleculares , Estructura Secundaria de Proteína , Rimantadina/química , Rimantadina/farmacología , Relación Estructura-Actividad , Proteínas Virales/química
6.
Drug Des Devel Ther ; 12: 1019-1031, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29750015

RESUMEN

BACKGROUND: Adamantane-based compounds have been identified to interfere with the ion-channel activity of viroporins and thereby inhibit viral infection. To better understand the difference in the inhibition mechanism of viroporins, we synthesized symmetric dimeric adamantane analogs of various alkyl-spacer lengths. METHODS: Symmetric dimeric adamantane derivatives were synthesized where two amantadine or rimantadine molecules were linked by various alkyl-spacers. The inhibitory activity of the compounds was studied on two viroporins: the influenza virus M2 protein, expressed in Xenopus oocytes, using the two-electrode voltage-clamp technique, and the hepatitis C virus (HCV) p7 channels for five different genotypes (1a, 1b, 2a, 3a, and 4a) expressed in HEK293 cells using whole-cell patch-clamp recording techniques. RESULTS: Upon testing on M2 protein, dimeric compounds showed significantly lower inhibitory activity relative to the monomeric amantadine. The lack of channel blockage of the dimeric amantadine and rimantadine analogs against M2 wild type and M2-S31N mutant was consistent with previously proposed drug-binding mechanisms and further confirmed that the pore-binding model is the pharmacologically relevant drug-binding model. On the other hand, these dimers showed similar potency to their respective monomeric analogs when tested on p7 protein in HCV genotypes 1a, 1b, and 4a while being 700-fold and 150-fold more potent than amantadine in genotypes 2a and 3a, respectively. An amino group appears to be important for inhibiting the ion-channel activity of p7 protein in genotype 2a, while its importance was minimal in all other genotypes. CONCLUSION: Symmetric dimeric adamantanes can be considered a prospective class of p7 inhibitors that are able to address the differences in adamantane sensitivity among the various genotypes of HCV.


Asunto(s)
Adamantano/farmacología , Amantadina/farmacología , Antivirales/farmacología , Rimantadina/farmacología , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas Virales/antagonistas & inhibidores , Adamantano/síntesis química , Adamantano/química , Amantadina/síntesis química , Amantadina/química , Antivirales/síntesis química , Antivirales/química , Células Cultivadas , Diseño de Fármacos , Células HEK293 , Humanos , Modelos Moleculares , Estructura Molecular , Rimantadina/síntesis química , Rimantadina/química , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/metabolismo
7.
Biochim Biophys Acta Gen Subj ; 1862(3): 692-704, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29221984

RESUMEN

BACKGROUND: In theory, binding enthalpies directly obtained from calorimetry (such as ITC) and the temperature dependence of the binding free energy (van't Hoff method) should agree. However, previous studies have often found them to be discrepant. METHODS: Experimental binding enthalpies (both calorimetric and van't Hoff) are obtained for two host-guest pairs using ITC, and the discrepancy between the two enthalpies is examined. Modeling of artificial ITC data is also used to examine how different sources of error propagate to both types of binding enthalpies. RESULTS: For the host-guest pairs examined here, good agreement, to within about 0.4kcal/mol, is obtained between the two enthalpies. Additionally, using artificial data, we find that different sources of error propagate to either enthalpy uniquely, with concentration error and heat error propagating primarily to calorimetric and van't Hoff enthalpies, respectively. CONCLUSIONS: With modern calorimeters, good agreement between van't Hoff and calorimetric enthalpies should be achievable, barring issues due to non-ideality or unanticipated measurement pathologies. Indeed, disagreement between the two can serve as a flag for error-prone datasets. A review of the underlying theory supports the expectation that these two quantities should be in agreement. GENERAL SIGNIFICANCE: We address and arguably resolve long-standing questions regarding the relationship between calorimetric and van't Hoff enthalpies. In addition, we show that comparison of these two quantities can be used as an internal consistency check of a calorimetry study.


Asunto(s)
Calorimetría/métodos , Termodinámica , Algoritmos , Amantadina/química , Calorimetría/instrumentación , Transferencia de Energía , Calor , Cinética , Rimantadina/química , beta-Ciclodextrinas/química
8.
J Am Chem Soc ; 138(5): 1506-9, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26804976

RESUMEN

Rimantadine hydrochloride (α-methyl-1-adamantane-methalamine hydrochloride) is a chiral compound which exerts antiviral activity against the influenza A virus by inhibiting proton conductance of the M2 ion channel. In complex with M2, rimantadine has always been characterized as a racemic mixture. Here, we report the novel enantioselective synthesis of deuterium-labeled (R)- and (S)-rimantadine and the characterization of their protein-ligand interactions using solid-state NMR. Isotropic chemical shift changes strongly support differential binding of the enantiomers to the proton channel. Position restrained simulations satisfying distance restraints from (13)C-(2)H rotational-echo double-resonance NMR show marked differences in the hydrogen-bonding pattern of the two enantiomers at the binding site. Together these results suggest a complex set of interactions between (R)-rimantadine and the M2 proton channel, leading to a higher stability for this enantiomer of the drug in the channel pore.


Asunto(s)
Antivirales/metabolismo , Rimantadina/metabolismo , Proteínas de la Matriz Viral/metabolismo , Antivirales/química , Enlace de Hidrógeno , Espectroscopía de Resonancia Magnética , Unión Proteica , Rimantadina/química , Estereoisomerismo
9.
Curr Med Chem ; 22(11): 1361-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25723505

RESUMEN

The annual flu season causes thousands of deaths and millions of hospitalizations, which pose a great burden to global health and economy. Moreover, a flu pandemic arising from reassortment viruses, such as H5N1 and H1N1, raises even greater concern due to the lack of effective vaccines at the initial stage of flu outbreak. The influenza virus is the causative agent of flu infection. Currently there are four drugs in use to combat influenza infection. Amantadine and rimantadine are M2 proton channel blockers that inhibit virus uncoating; oseltamivir and zanamivir are neuraminidase (NA) inhibitors that inhibit virus release. However, recent years have witnessed a drastic increase in instances of drug resistance, and flu strains that are resistant to both classes of drugs have been reported. Thus, there is a pressing need to develop the next generation of anti-influenza drugs. Among a handful of anti-influenza drug targets, the viral fusion protein hemagglutinin (HA) is one of the most advanced. This review discusses the biological roles of HA during viral replication and highlights peptide- and small molecule-based HA inhibitors, including recent computationally designed HA binders. The text is organized into four sections based on the maturation stages of HA: inhibitors targeting the glycosylation of HA, the proteolytic activation of HA, the attachment of HA to host cell receptors, and peptide- and small molecule-based inhibitors targeting HA-mediated membrane fusion. Of particular interest are advances in the areas of developing dual inhibitors targeting both HA and NA and broad-spectrum HA inhibitors targeting both groups of HAs.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Orthomyxoviridae/efectos de los fármacos , Amantadina/química , Amantadina/farmacología , Antivirales/química , Inhibidores Enzimáticos/química , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/metabolismo , Orthomyxoviridae/enzimología , Orthomyxoviridae/metabolismo , Oseltamivir/química , Oseltamivir/farmacología , Rimantadina/química , Rimantadina/farmacología , Zanamivir/química , Zanamivir/farmacología
10.
J Pharm Sci ; 104(4): 1561-2, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25583633

RESUMEN

The utility of cyclodextrin (CD) complexation in improving apparent solubility of drugs in parenteral formulations is well established. Administration of these formulations delivers CD directly into the systemic circulation, and it may be necessary to demonstrate unaltered in vivo disposition of a drug coadministered with a CD. Crucial to the undertaking of such a study is the need for bioanalytical assays in which CD presence does not impact drug quantitation. This is of particular importance when assessing the potential impact of in vivo CD complexation on the urinary excretion of a drug, as CDs are predominantly eliminated via glomerular filtration, and hence are present in urine at significantly higher concentration than would be present in blood and plasma. Of 23 publications (in the past 30 years) describing preclinical and clinical assessment of drug pharmacokinetics after i.v. administration of CD-enabled formulations, only two reports clearly stated that the presence of CD had no impact on assay performance. In this work, we describe the simple process involved in (1) predicting the maximum concentrations of a modified CD, sulfobutylether7 -ß-CD (SBE7 -ß-CD), in plasma and urine samples from preclinical studies, and (2) evaluating the impact of SBE7 -ß-CD on the quantitative liquid chromatography-mass spectrometry analysis of rimantadine.


Asunto(s)
Cromatografía Liquida , Excipientes/administración & dosificación , Excipientes/farmacocinética , Espectrometría de Masas , Rimantadina/farmacocinética , Tecnología Farmacéutica/métodos , beta-Ciclodextrinas/farmacocinética , Administración Intravenosa , Animales , Calibración , Química Farmacéutica , Cromatografía Liquida/normas , Excipientes/metabolismo , Espectrometría de Masas/normas , Modelos Biológicos , Ratas , Estándares de Referencia , Reproducibilidad de los Resultados , Rimantadina/administración & dosificación , Rimantadina/sangre , Rimantadina/química , Rimantadina/orina , Tecnología Farmacéutica/normas , beta-Ciclodextrinas/administración & dosificación , beta-Ciclodextrinas/sangre , beta-Ciclodextrinas/química , beta-Ciclodextrinas/orina
11.
Fundam Clin Pharmacol ; 28(2): 199-204, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23231589

RESUMEN

The aim of this study was to investigate infiltrative cutaneous anesthesia of 2-adamantanamine and rimantadine. After subcutaneous injections of drugs in rats, the blockade of cutaneous trunci muscle reflex by 2-adamantanamine and rimantadine was evaluated. Lidocaine, a common local anesthetic, was used as control. We showed that rimantadine and 2-adamantanamine as well as the local anesthetic lidocaine produced infiltrative anesthesia of skin in a dose-dependent fashion. Saline (vehicle) group displayed no cutaneous anesthesia. The relative potency of these drugs was rimantadine [23.8 (21.1-26.8)] = lidocaine [26.4 (22.7-30.6)] > 2-adamantanamine [64.6 (55.0-75.9)] (P < 0.01). On an equianesthetic basis [25% effective dose (ED25 ), ED50 , and ED75 ], rimantadine and 2-adamantanamine had longer duration of action than lidocaine (P < 0.05). Neither local injection of saline nor intraperitoneal administration of a large dose of drugs elicited cutaneous anesthesia (data not shown). These data demonstrated for the first time that rimantadine had a similar potent and longer duration of skin infiltrative anesthesia than did lidocaine, whereas 2-adamantanamine had a less potency but longer duration of cutaneous anesthesia than did lidocaine.


Asunto(s)
Amantadina/análogos & derivados , Anestesia Local/métodos , Anestésicos Locales/farmacología , Rimantadina/farmacología , Piel/efectos de los fármacos , Amantadina/administración & dosificación , Amantadina/química , Amantadina/farmacología , Anestésicos Locales/administración & dosificación , Anestésicos Locales/química , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inyecciones Subcutáneas , Masculino , Estructura Molecular , Dimensión del Dolor , Estimulación Física , Ratas , Ratas Sprague-Dawley , Reflejo/efectos de los fármacos , Rimantadina/administración & dosificación , Rimantadina/química , Piel/inervación
12.
J Pharm Sci ; 102(8): 2450-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23832466

RESUMEN

By attaching multiple copies of the influenza M2 ion channel inhibitors amantadine (1) and rimantadine (2) to polymeric chains, we endeavored to recover their potency in inhibiting drug-resistant influenza viruses. Depending on loading densities, as well as the nature of the drug, the polymer, and the spacer arm, polymer-conjugated drugs were up to 30-fold more potent inhibitors of drug-resistant strains than their monomeric parents. In particular, a 20% loading density and a short linker group on the negatively charged poly-l-glutamate resulted in one of the most potent inhibitors for 2's conjugates against drug-resistant influenza strains. Although full recovery of the inhibitory action against drug-resistant strains was not achieved, this study may be a step toward salvaging anti-influenza drugs that are no longer effective.


Asunto(s)
Amantadina/administración & dosificación , Antivirales/administración & dosificación , Portadores de Fármacos/química , Virus de la Influenza A/efectos de los fármacos , Polímeros/química , Rimantadina/administración & dosificación , Amantadina/química , Amantadina/farmacología , Animales , Antivirales/química , Antivirales/farmacología , Línea Celular , Perros , Farmacorresistencia Viral , Humanos , Gripe Humana/tratamiento farmacológico , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Ácido Poliglutámico/química , Rimantadina/química , Rimantadina/farmacología
13.
Nature ; 498(7455): 521-5, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23739335

RESUMEN

The hepatitis C virus (HCV) has developed a small membrane protein, p7, which remarkably can self-assemble into a large channel complex that selectively conducts cations. We wanted to examine the structural solution that the viroporin adopts in order to achieve selective cation conduction, because p7 has no homology with any of the known prokaryotic or eukaryotic channel proteins. The activity of p7 can be inhibited by amantadine and rimantadine, which are potent blockers of the influenza M2 channel and licensed drugs against influenza infections. The adamantane derivatives have been used in HCV clinical trials, but large variation in drug efficacy among the various HCV genotypes has been difficult to explain without detailed molecular structures. Here we determine the structures of this HCV viroporin as well as its drug-binding site using the latest nuclear magnetic resonance (NMR) technologies. The structure exhibits an unusual mode of hexameric assembly, where the individual p7 monomers, i, not only interact with their immediate neighbours, but also reach farther to associate with the i+2 and i+3 monomers, forming a sophisticated, funnel-like architecture. The structure also points to a mechanism of cation selection: an asparagine/histidine ring that constricts the narrow end of the funnel serves as a broad cation selectivity filter, whereas an arginine/lysine ring that defines the wide end of the funnel may selectively allow cation diffusion into the channel. Our functional investigation using whole-cell channel recording shows that these residues are critical for channel activity. NMR measurements of the channel-drug complex revealed six equivalent hydrophobic pockets between the peripheral and pore-forming helices to which amantadine or rimantadine binds, and compound binding specifically to this position may allosterically inhibit cation conduction by preventing the channel from opening. Our data provide a molecular explanation for p7-mediated cation conductance and its inhibition by adamantane derivatives.


Asunto(s)
Hepacivirus/química , Proteínas Virales/química , Adamantano/análogos & derivados , Adamantano/química , Adamantano/metabolismo , Adamantano/farmacología , Sitios de Unión , Difusión , Microscopía Electrónica , Modelos Biológicos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Porosidad , Rimantadina/química , Rimantadina/metabolismo , Rimantadina/farmacología , Relación Estructura-Actividad , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura
14.
Biochemistry ; 52(16): 2774-82, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23480101

RESUMEN

We demonstrate the use of dynamic nuclear polarization (DNP) to elucidate ligand binding to a membrane protein using dipolar recoupling magic angle spinning (MAS) NMR. In particular, we detect drug binding in the proton transporter M2(18-60) from influenza A using recoupling experiments at room temperature and with cryogenic DNP. The results indicate that the pore binding site of rimantadine is correlated with previously reported widespread chemical shift changes, suggesting functional binding in the pore. Futhermore, the (15)N-labeled ammonium of rimantadine was observed near A30 (13)Cß and G34 (13)Cα, suggesting a possible hydrogen bond to A30 carbonyl. Cryogenic DNP was required to observe the weaker external binding site(s) in a ZF-TEDOR spectrum. This approach is generally applicable, particularly for weakly bound ligands, in which case the application of MAS NMR dipolar recoupling requires the low temperatures to quench dynamic exchange processes. For the fully protonated samples investigated, we observed DNP signal enhancements of ~10 at 400 MHz using only 4-6 mM of the polarizing agent TOTAPOL. At 600 MHz and with DNP, we measured a distance between the drug and the protein to a precision of 0.2 Å.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Rimantadina/metabolismo , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/metabolismo , Secuencia de Aminoácidos , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Sitios de Unión , Frío , Glicerol/química , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Isótopos de Nitrógeno , Conformación Proteica , Rimantadina/química , Rimantadina/farmacología , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-22257715

RESUMEN

A novel gold(I) complex with rimantadine (RTD) was obtained and structurally characterized by a set of chemical and spectroscopic analysis. 1H, 13C and 15N nuclear magnetic resonance (NMR) and infrared (IR) spectroscopic measurements suggest coordination of the ligand to Au(I) through the N atom of the ethanamine group. Theoretical (DFT) calculations confirmed the IR assignments and permit proposing an optimized geometry for the complex. The gold(I)-rimantadine complex (Au-RTD) is soluble in methanol, ethanol, dimethylsulfoxide, acetone and acetonitrile. The preliminary kinetic studies based on UV-vis spectroscopic measurements indicate the stability of the compound in solution. Antibacterial activities of the complex were evaluated by an antibiogram assay. The Au-RTD complex showed an effective in vitro antibacterial activity against the Pseudomonas aeruginosa, Escherichia coli (Gram-negative), and Staphylococcus aureus (Gram-positive) bacterial strains.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Compuestos Orgánicos de Oro/química , Compuestos Orgánicos de Oro/farmacología , Rimantadina/química , Rimantadina/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Escherichia coli/efectos de los fármacos , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Pseudomonas aeruginosa/efectos de los fármacos , Espectrofotometría Infrarroja , Staphylococcus aureus/efectos de los fármacos
16.
Molecules ; 16(12): 10227-55, 2011 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-22158591

RESUMEN

The M2 channel protein on the influenza A virus membrane has become the main target of the anti-flu drugs amantadine and rimantadine. The structure of the M2 channel proteins of the H3N2 (PDB code 2RLF) and 2009-H1N1 (Genbank accession number GQ385383) viruses may help researchers to solve the drug-resistant problem of these two adamantane-based drugs and develop more powerful new drugs against influenza A virus. In the present study, we searched for new M2 channel inhibitors through a combination of different computational methodologies, including virtual screening with docking and pharmacophore modeling. Virtual screening was performed to calculate the free energies of binding between receptor M2 channel proteins and 200 new designed ligands. After that, pharmacophore analysis was used to identify the important M2 protein-inhibitor interactions and common features of top binding compounds with M2 channel proteins. Finally, the two most potential compounds were determined as novel leads to inhibit M2 channel proteins in both H3N2 and 2009-H1N1 influenza A virus.


Asunto(s)
Amantadina/química , Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Moduladores del Transporte de Membrana/farmacología , Modelos Moleculares , Interfaz Usuario-Computador , Proteínas de la Matriz Viral/antagonistas & inhibidores , Antivirales/química , Sitios de Unión , Enlace de Hidrógeno/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Moduladores del Transporte de Membrana/química , Rimantadina/química , Relación Estructura-Actividad , Termodinámica , Proteínas de la Matriz Viral/química
17.
Structure ; 19(11): 1655-63, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22078564

RESUMEN

The M2 channel of influenza A is a target of the adamantane family antiviral drugs. Two different drug-binding sites have been reported: one inside the pore, and the other is a lipid-facing pocket. A previous study showed that a chimera of M2 variants from influenza A and B that contains only the pore-binding site is sensitive to amantadine inhibition, suggesting that the primary site of inhibition is inside the pore. To obtain atomic details of channel-drug interaction, we determined the structures of the chimeric channel with and without rimantadine. Inside the channel and near the N-terminal end, methyl groups of Val27 and Ala30 from four subunits form a hydrophobic pocket around the adamantane, and the drug amino group appears to be in polar contact with the backbone oxygen of Ala30. The structures also reveal differences between the drug-bound and -unbound states of the channel that can explain drug resistance.


Asunto(s)
Antivirales/química , Virus de la Influenza A , Rimantadina/química , Proteínas Virales de Fusión/química , Secuencias de Aminoácidos , Sitios de Unión , Liposomas/química , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Unión Proteica , Estructura Cuaternaria de Proteína , Proteínas Virales de Fusión/antagonistas & inhibidores , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/química
18.
Protein Eng Des Sel ; 23(8): 663-6, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20571121

RESUMEN

As an essential component of the viral envelope, M2 proton channel plays a central role in the virus replications and has been a key target for drug design against the influenza A viruses. The adamantadine-based drugs, such as amantadine and rimantadine, were developed for blocking the channel so as to suppress the replication of viruses. However, patients, especially those infected by the H1N1 influenza A viruses, are increasingly suffering from the drug-resistance problem. According to the findings revealed recently by the high-resolution NMR studies, the drug-resistance problem is due to the structural allostery caused by some mutations, such as L26F, V27A and S31N, in the four-helix bundle of the channel. In this study, we are to address this problem from a dynamic point of view by conducting molecular dynamics (MD) simulations on both the open and the closed states of the wild-type (WT) and S31N mutant M2 channels in the presence of rimantadine. It was observed from the MD simulated structures that the mutant channel could still keep open even if binding with rimantadine, but the WT channel could not. This was because the mutation would destabilize the helix bundle and trigger it from a compact packing state to a loose one. It is anticipated that the findings may provide useful insights for in-depth understanding the action mechanism of the M2 channel and developing more-effective drugs against influenza A viruses.


Asunto(s)
Proteínas de la Matriz Viral/química , Farmacorresistencia Viral , Simulación de Dinámica Molecular , Mutación , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Rimantadina/química , Rimantadina/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
19.
PLoS One ; 5(2): e9388, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20186344

RESUMEN

BACKGROUND: M2 proton channel of H1N1 influenza A virus is the target protein of anti-flu drugs amantadine and rimantadine. However, the two once powerful adamantane-based drugs lost their 90% bioactivity because of mutations of virus in recent twenty years. The NMR structure of the M2 channel protein determined by Schnell and Chou (Nature, 2008, 451, 591-595) may help people to solve the drug-resistant problem and develop more powerful new drugs against H1N1 influenza virus. METHODOLOGY: Docking calculation is performed to build the complex structure between receptor M2 proton channel and ligands, including existing drugs amantadine and rimantadine, and two newly designed inhibitors. The computer-aided drug design methods are used to calculate the binding free energies, with the computational biology techniques to analyze the interactions between M2 proton channel and adamantine-based inhibitors. CONCLUSIONS: 1) The NMR structure of M2 proton channel provides a reliable structural basis for rational drug design against influenza virus. 2) The channel gating mechanism and the inhibiting mechanism of M2 proton channel, revealed by the NMR structure of M2 proton channel, provides the new ideas for channel inhibitor design. 3) The newly designed adamantane-based inhibitors based on the modeled structure of H1N1-M2 proton channel have two pharmacophore groups, which act like a "barrel hoop", holding two adjacent helices of the H1N1-M2 tetramer through the two pharmacophore groups outside the channel. 4) The inhibitors with such binding mechanism may overcome the drug resistance problem of influenza A virus to the adamantane-based drugs.


Asunto(s)
Antivirales/química , Diseño de Fármacos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Proteínas de la Matriz Viral/química , Adamantano/química , Adamantano/farmacología , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Sitios de Unión/genética , Farmacorresistencia Viral/genética , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/genética , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/química , Canales Iónicos/genética , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutación , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Rimantadina/química , Rimantadina/farmacología , Homología de Secuencia de Aminoácido , Porcinos , Enfermedades de los Porcinos/virología , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/genética
20.
Yao Xue Xue Bao ; 45(3): 289-99, 2010 Mar.
Artículo en Chino | MEDLINE | ID: mdl-21351503

RESUMEN

Influenza is a major threat to millions of people worldwide. Vaccines and antiviral agents are two main options available to reduce the impact of the influenza virus, while anti-influenza agents are the most effective means to prevent the transmission of the highly contagious virus and to treat the epidemics of disease. At present, four anti-influenza agents have been approved by the FDA for the treatment of influenza, including two M2 protein ion channel inhibitors-amantadine and rimantadine and two neuraminidase inhibitors-zanamivir and oseltamivir. Arbidol hydrochloride, launched in Russia, is a potent inhibitor of influenza virus, too. Neuraminidase inhibitors could be classified generally by structure into six different kinds: sialic acid derivatives, benzoic acid derivatives, cyclohexene derivatives, cyclopentane derivatives, pyrrolidine derivatives and natural products. In this paper, recent progress in the research of the action mechanisms and structure-activity relationships of these anti-influenza virus agents were reviewed.


Asunto(s)
Antivirales , Neuraminidasa/antagonistas & inhibidores , Orthomyxoviridae/efectos de los fármacos , Proteínas de la Matriz Viral/antagonistas & inhibidores , Ácidos Carbocíclicos , Amantadina/síntesis química , Amantadina/química , Amantadina/farmacología , Amantadina/uso terapéutico , Antivirales/síntesis química , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , Ciclopentanos/síntesis química , Ciclopentanos/química , Ciclopentanos/farmacología , Ciclopentanos/uso terapéutico , Guanidinas/síntesis química , Guanidinas/química , Guanidinas/farmacología , Guanidinas/uso terapéutico , Humanos , Indoles/síntesis química , Indoles/química , Indoles/farmacología , Indoles/uso terapéutico , Gripe Humana/tratamiento farmacológico , Neuraminidasa/síntesis química , Neuraminidasa/química , Neuraminidasa/farmacología , Neuraminidasa/uso terapéutico , Oseltamivir/síntesis química , Oseltamivir/química , Oseltamivir/farmacología , Oseltamivir/uso terapéutico , Pirrolidinas/síntesis química , Pirrolidinas/química , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , Rimantadina/síntesis química , Rimantadina/química , Rimantadina/farmacología , Rimantadina/uso terapéutico , Relación Estructura-Actividad , Proteínas de la Matriz Viral/síntesis química , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/farmacología , Proteínas de la Matriz Viral/uso terapéutico , Zanamivir/síntesis química , Zanamivir/química , Zanamivir/farmacología , Zanamivir/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...