Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Viruses ; 12(2)2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32033244

RESUMEN

The green tea catechin epigallocatechin gallate (EGCG) exhibits antiviral activity against various viruses. Whether EGCG also inhibits the infectivity of circovirus remains unclear. In this study, we demonstrated the antiviral effect of EGCG on porcine circovirus type 2 (PCV2). EGCG targets PCV2 virions directly and blocks the attachment of virions to host cells. The microscale thermophoresis assay showed EGCG could interact with PCV2 capsid protein in vitro with considerable affinity (Kd = 98.03 ± 4.76 µM), thereby interfering with the binding of the capsid to the cell surface receptor heparan sulfate. The molecular docking analysis of capsid-EGCG interaction identified the key amino acids which formed the binding pocket accommodating EGCG. Amino acids ARG51, ASP70, ARG73 and ASP78 of capsid were found to be critical for maintaining the binding, and the arginine residues were also essential for the electrostatic interaction with heparan sulfate. The rescued mutant viruses also confirm the importance of the key amino acids of the capsid to the antiviral effect of EGCG. Our findings suggest that catechins could act as anti-infective agents against circovirus invasion, as well as provide the basic information for the development and synthesis of structure-based anti-circovirus drugs.


Asunto(s)
Antivirales/farmacología , Cápside/metabolismo , Catequina/análogos & derivados , Circovirus/efectos de los fármacos , Acoplamiento Viral/efectos de los fármacos , Animales , Cápside/química , Cápside/efectos de los fármacos , Catequina/farmacología , Línea Celular , Circovirus/clasificación , Simulación del Acoplamiento Molecular , Porcinos , Té/química
2.
Antiviral Res ; 169: 104544, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31254557

RESUMEN

Due to its multifaceted essential roles in virus replication and extreme genetic fragility, the human immunodeficiency virus type 1 (HIV-1) capsid (CA) protein is a valued therapeutic target. However, CA is as yet unexploited clinically, as there are no antiviral agents that target it currently on the market. To facilitate the identification of potential HIV-1 CA inhibitors, we established a homogeneous time-resolved fluorescence (HTRF) assay to screen for small molecules that target a biologically active and specific binding pocket in the C-terminal domain of HIV-1 CA (CA CTD). The assay, which is based on competition of small molecules for the binding of a known CA inhibitor (CAI) to the CA CTD, exhibited a signal-to-background ratio (S/B) > 10 and a Z' value > 0.9. In a pilot screen of three kinase inhibitor libraries containing 464 compounds, we identified one compound, TX-1918, as a low micromolecular inhibitor of the HIV-1 CA CTD-CAI interaction (IC50 = 3.81 µM) that also inhibited viral replication at moderate micromolar concentration (EC50 = 15.16 µM) and inhibited CA assembly in vitro. Based on the structure of TX-1918, an additional compound with an antiviral EC50 of 6.57 µM and cellular cytotoxicity CC50 of 102.55 µM was obtained from a compound similarity search. Thus, the HTRF-based assay has properties that are suitable for screening large compound libraries to identify novel anti-HIV-1 inhibitors targeting the CA CTD.


Asunto(s)
Unión Competitiva , Proteínas de la Cápside/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Fluorescencia , VIH-1/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Ensamble de Virus/efectos de los fármacos , Cápside/efectos de los fármacos , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Liberación de Fármacos , Proteínas Recombinantes , Linfocitos T , Replicación Viral/efectos de los fármacos
3.
Nat Commun ; 10(1): 2184, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31097716

RESUMEN

Chronic hepatitis B virus (HBV) infection can cause cirrhosis and hepatocellular carcinoma and is therefore a serious public health problem. Infected patients are currently treated with nucleoside/nucleotide analogs and interferon α, but this approach is not curative. Here, we screen 978 FDA-approved compounds for their ability to inhibit HBV replication in HBV-expressing HepG2.2.15 cells. We find that ciclopirox, a synthetic antifungal agent, strongly inhibits HBV replication in cells and in mice by blocking HBV capsid assembly. The crystal structure of the HBV core protein and ciclopirox complex reveals a unique binding mode at dimer-dimer interfaces. Ciclopirox synergizes with nucleoside/nucleotide analogs to prevent HBV replication in cells and in a humanized liver mouse model. Therefore, orally-administered ciclopirox may provide a novel opportunity to combat chronic HBV infection by blocking HBV capsid assembly.


Asunto(s)
Antivirales/farmacología , Ciclopirox/farmacología , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/tratamiento farmacológico , Ensamble de Virus/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Cápside/efectos de los fármacos , Cápside/metabolismo , Ciclopirox/química , Ciclopirox/uso terapéutico , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Células Hep G2 , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/patología , Hepatitis B Crónica/virología , Hepatocitos/trasplante , Hepatocitos/virología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , ARN Viral/metabolismo , Quimera por Trasplante , Resultado del Tratamiento , Proteínas del Núcleo Viral/química , Proteínas del Núcleo Viral/metabolismo , Replicación Viral/efectos de los fármacos
4.
Artículo en Inglés | MEDLINE | ID: mdl-30373799

RESUMEN

NVR 3-778 is the first capsid assembly modulator (CAM) that has demonstrated antiviral activity in hepatitis B virus (HBV)-infected patients. NVR 3-778 inhibited the generation of infectious HBV DNA-containing virus particles with a mean antiviral 50% effective concentration (EC50) of 0.40 µM in HepG2.2.15 cells. The antiviral profile of NVR 3-778 indicates pan-genotypic antiviral activity and a lack of cross-resistance with nucleos(t)ide inhibitors of HBV replication. The combination of NVR 3-778 with nucleos(t)ide analogs in vitro resulted in additive or synergistic antiviral activity. Mutations within the hydrophobic pocket at the dimer-dimer interface of the core protein could confer resistance to NVR 3-778, which is consistent with the ability of the compound to bind to core and to induce capsid assembly. By targeting core, NVR 3-778 inhibits pregenomic RNA encapsidation, viral replication, and the production of HBV DNA- and HBV RNA-containing particles. NVR 3-778 also inhibited de novo infection and viral replication in primary human hepatocytes with EC50 values of 0.81 µM against HBV DNA and between 3.7 and 4.8 µM against the production of HBV antigens and intracellular HBV RNA. NVR 3-778 showed favorable pharmacokinetics and safety in animal species, allowing serum levels in excess of 100 µM to be achieved in mice and, thus, enabling efficacy studies in vivo The overall preclinical profile of NVR 3-778 predicts antiviral activity in vivo and supports its further evaluation for safety, pharmacokinetics, and antiviral activity in HBV-infected patients.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Cápside/efectos de los fármacos , ADN Viral/antagonistas & inhibidores , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Piperidinas/farmacología , ARN Viral/antagonistas & inhibidores , Animales , Antígenos Virales/genética , Antígenos Virales/metabolismo , Antivirales/sangre , Antivirales/química , Antivirales/farmacocinética , Benzamidas/sangre , Benzamidas/química , Benzamidas/farmacocinética , Cápside/química , Cápside/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Células Hep G2 , Hepatitis B/virología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Hepatocitos/virología , Humanos , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Piperidinas/sangre , Piperidinas/química , Piperidinas/farmacocinética , Cultivo Primario de Células , ARN Viral/genética , ARN Viral/metabolismo , Proteínas del Núcleo Viral/antagonistas & inhibidores , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Replicación Viral/efectos de los fármacos
5.
Biotechnol Adv ; 36(3): 557-576, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29292156

RESUMEN

Despite successful vaccination programs and effective treatments for some viral infections, humans are still losing the battle with viruses. Persisting human pandemics, emerging and re-emerging viruses, and evolution of drug-resistant strains impose continuous search for new antiviral drugs. A combination of detailed information about the molecular organization of viruses and progress in molecular biology and computer technologies has enabled rational antivirals design. Initial step in establishing efficacy of new antivirals is based on simple methods assessing inhibition of the intended target. We provide here an overview of biochemical and cell-based assays evaluating the activity of inhibitors of clinically important viruses.


Asunto(s)
Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Fenómenos Fisiológicos de los Virus/efectos de los fármacos , Animales , Cápside/efectos de los fármacos , Cápside/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
6.
J Gen Virol ; 98(3): 385-395, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27902359

RESUMEN

The foot-and-mouth disease virus (FMDV) capsid precursor, P1-2A, is cleaved by FMDV 3C protease to yield VP0, VP3, VP1 and 2A. Cleavage of the VP1/2A junction is the slowest. Serotype O FMDVs with uncleaved VP1-2A (having a K210E substitution in VP1; at position P2 in cleavage site) have been described previously and acquired a second site substitution (VP1 E83K) during virus rescue. Furthermore, introduction of the VP1 E83K substitution alone generated a second site change at the VP1/2A junction (2A L2P, position P2' in cleavage site). These virus adaptations have now been analysed using next-generation sequencing to determine sub-consensus level changes in the virus; this revealed other variants within the E83K mutant virus population that changed residue VP1 K210. The construction of serotype A viruses with a blocked VP1/2A cleavage site (containing K210E) has now been achieved. A collection of alternative amino acid substitutions was made at this site, and the properties of the mutant viruses were determined. Only the presence of a positively charged residue at position P2 in the cleavage site permitted efficient cleavage of the VP1/2A junction, consistent with analyses of diverse FMDV genome sequences. Interestingly, in contrast to the serotype O virus results, no second site mutations occurred within the VP1 coding region of serotype A viruses with the blocked VP1/2A cleavage site. However, some of these viruses acquired changes in the 2C protein that is involved in enterovirus morphogenesis. These results have implications for the testing of potential antiviral agents targeting the FMDV 3C protease.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Cisteína Endopeptidasas/metabolismo , Virus de la Fiebre Aftosa/metabolismo , Fiebre Aftosa/virología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Sustitución de Aminoácidos , Animales , Antivirales/farmacología , Cápside/efectos de los fármacos , Proteínas de la Cápside/genética , Evaluación Preclínica de Medicamentos , Virus de la Fiebre Aftosa/efectos de los fármacos , Virus de la Fiebre Aftosa/genética , Ácido Glutámico/genética , Lisina/genética , Mutación , Ensamble de Virus/efectos de los fármacos
7.
J Antimicrob Chemother ; 71(7): 1922-32, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27098012

RESUMEN

OBJECTIVES: Enterovirus 71 (EV-A71) is an important pathogen that can cause severe neurological symptoms and even death. Our aim was to identify potent anti-EV-A71 compounds and study their underlying mechanisms and in vivo activity. METHODS: We identified a potent imidazolidinone derivative (abbreviated to PR66) as an inhibitor of EV-A71 infection from the screening of compounds and subsequent structure-based modification. Time-course treatments and resistant virus selection of PR66 were employed to study the mode of mechanism of PR66. In vivo activity of PR66 was tested in the ICR strain of new-born mice challenged with EV-A71/4643/MP4. RESULTS: PR66 could impede the uncoating process during viral infection via interaction with capsid protein VP1, as shown by a resistant virus selection assay. Using site-directed mutagenesis, we confirmed that a change from valine to phenylalanine in the 179th amino acid residue of the cDNA-derived resistant virus resulted in resistance to PR66. PR66 increased the virion stability of WT viruses, but not the PR66-resistant mutant, in a particle stability thermal release assay. We further showed that PR66 had excellent anti-EV-A71 activity in an in vivo mouse model of disease, with a dose-dependent increase in survival rate and in protection against virus-induced hind-limb paralysis following oral or intraperitoneal administration. This was associated with reductions of viral titres in brain and muscle tissues. CONCLUSIONS: We demonstrated here for the first time that an imidazolidinone derivative (PR66) could protect against EV-A71-induced neurological symptoms in vivo by suppressing EV-A71 replication. This involved binding to and restricting viral uncoating.


Asunto(s)
Antivirales/metabolismo , Antivirales/farmacología , Cápside/efectos de los fármacos , Enterovirus Humano A/efectos de los fármacos , Animales , Antivirales/aislamiento & purificación , Línea Celular , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/virología , Humanos , Concentración 50 Inhibidora , Ratones Endogámicos ICR , Análisis de Supervivencia
8.
J Virol ; 89(10): 5350-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25741002

RESUMEN

UNLABELLED: During uncoating, the conical capsid of HIV disassembles by dissociation of the p24 capsid protein (CA). Uncoating is known to be required for HIV replication, but the mechanism is poorly defined. Here, we examined the timing and effect of two capsid binding drugs (PF74 and BI2) on infectivity and capsid integrity in HIV-1-infected cells. The virus remained susceptible to the action of PF74 and BI2 for hours after uncoating as defined in parallel drug addition and cyclosporine (CsA) washout assays to detect the kinetics of drug susceptibility and uncoating, respectively. Resistance mutations in CA decreased the potency of these compounds, demonstrating that CA is the target of drug action. However, neither drug altered capsid integrity in a fluorescence microscopy-based assay. These data suggest that PF74 and BI2 do not alter HIV-1 uncoating but rather affect a later step in viral replication. Because both drugs bind CA, we hypothesized that a residual amount of CA associates with the viral complex after the loss of the conical capsid to serve as a target for these drugs. Superresolution structured illumination microscopy (SIM) revealed that CA localized to viral complexes in the nuclei of infected cells. Using image quantification, we determined that viral complexes localized in the nucleus displayed a smaller amount of CA than complexes at the nuclear membrane, in the cytoplasm, or in controls. Collectively, these data suggest that a subset of CA remains associated with the viral complex after uncoating and that this residual CA is the target of PF74 and BI2. IMPORTANCE: The HIV-1 capsid is a target of interest for new antiviral therapies. This conical capsid is composed of monomers of the viral CA protein. During HIV-1 replication, the capsid must disassemble by a poorly defined process called uncoating. CA has also been implicated in later steps of replication, including nuclear import and integration. In this study, we used cell-based assays to examine the effect of two CA binding drugs (PF74 and BI2) on viral replication in infected cells. HIV-1 was susceptible to both drugs for hours after uncoating, suggesting that these drugs affect later steps of viral replication. High-resolution structured illumination microscopy (SIM) revealed that a subset of CA localized to viral complexes in the nuclei of cells. Collectively, these data suggest that a subset of CA remains associated with the viral complex after uncoating, which may facilitate later steps of viral replication and serve as a drug target.


Asunto(s)
Proteína p24 del Núcleo del VIH/fisiología , VIH-1/fisiología , Desencapsidación Viral/fisiología , Fármacos Anti-VIH/farmacología , Cápside/efectos de los fármacos , Cápside/fisiología , Línea Celular , Núcleo Celular/virología , Células HEK293 , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Células HeLa , Humanos , Indoles/farmacología , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología , Desencapsidación Viral/efectos de los fármacos
9.
Elife ; 32014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25365453

RESUMEN

The emergence of drug resistance can defeat the successful treatment of pathogens that display high mutation rates, as exemplified by RNA viruses. Here we detail a new paradigm in which a single compound directed against a 'dominant drug target' suppresses the emergence of naturally occurring drug-resistant variants in mice and cultured cells. All new drug-resistant viruses arise during intracellular replication and initially express their phenotypes in the presence of drug-susceptible genomes. For the targets of most anti-viral compounds, the presence of these drug-susceptible viral genomes does not prevent the selection of drug resistance. Here we show that, for an inhibitor of the function of oligomeric capsid proteins of poliovirus, the expression of drug-susceptible genomes causes chimeric oligomers to form, thus rendering the drug-susceptible genomes dominant. The use of dominant drug targets should suppress drug resistance whenever multiple genomes arise in the same cell and express products in a common milieu.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/efectos de los fármacos , Poliovirus/fisiología , Animales , Antivirales/uso terapéutico , Cápside/efectos de los fármacos , Cápside/metabolismo , Evaluación Preclínica de Medicamentos , Genoma Viral , Guanidina/farmacología , Guanidina/uso terapéutico , Células HeLa , Humanos , Ratones , Poliomielitis/tratamiento farmacológico , Poliomielitis/virología , Poliovirus/efectos de los fármacos , Poliovirus/genética , Proteínas Virales/metabolismo , Virión/efectos de los fármacos , Virión/metabolismo
10.
Biomaterials ; 34(17): 4203-13, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23498895

RESUMEN

Materials that interact in a controlled manner with viruses attract increasing interest in biotechnology, medicine, and environmental technology. Here, we show that virus-material interactions can be guided by intrinsic material surface chemistries, introduced by tailored surface functionalizations. For this purpose, colloidal alumina particles are surface functionalized with amino, carboxyl, phosphate, chloropropyl, and sulfonate groups in different surface concentrations and characterized in terms of elemental composition, electrokinetic, hydrophobic properties, and morphology. The interaction of the functionalized particles with hepatitis A virus and phages MS2 and PhiX174 is assessed by virus titer reduction after incubation with particles, activity of viruses conjugated to particles, and imaged by electron microscopy. Type and surface density of particle functional groups control the virus titer reduction between 0 and 99.999% (5 log values). For instance, high sulfonate surface concentrations (4.7 groups/nm(2)) inhibit attractive virus-material interactions and lead to complete virus recovery. Low sulfonate surface concentrations (1.2 groups/nm(2)), native alumina, and chloropropyl-functionalized particles induce strong virus-particle adsorption. The virus conformation and capsid amino acid composition further influence the virus-material interaction. Fundamental interrelations between material properties, virus properties, and the complex virus-material interaction are discussed and a versatile pool of surface functionalization strategies controlling virus-material interactions is presented.


Asunto(s)
Óxido de Aluminio/farmacología , Bacteriófago phi X 174/efectos de los fármacos , Coloides/farmacología , Levivirus/ultraestructura , Adsorción/efectos de los fármacos , Animales , Bacteriófago phi X 174/ultraestructura , Cápside/química , Cápside/efectos de los fármacos , Línea Celular , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Levivirus/efectos de los fármacos , Microscopía Electrónica de Transmisión , Electricidad Estática , Vapor , Propiedades de Superficie , Temperatura
11.
Nat Protoc ; 2(3): 490-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17406612

RESUMEN

Protein self-assembly is critical for numerous biological processes. Yet, assembly is rarely targeted by therapeutic agents, in part because it is hard to identify molecules that interfere with protein-protein interactions. Here, we describe a simple fluorescence-based screen for self-association and its application to the assembly of hepatitis B virus capsids. These data are analyzed to identify kinetic and thermodynamic effects--both of which are critical for the viral lifecycle and for understanding the mechanism of assembly effectors. Suggestions are made for modification of this protocol so that it can be applied to other self-assembling systems. With manual pipetting, setting up a plate takes about 2 h, the initial reading takes 1 h and the end point reading the following day takes about 5 min.


Asunto(s)
Antivirales/farmacología , Cápside/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Virus de la Hepatitis B/efectos de los fármacos , Ensamble de Virus/efectos de los fármacos , Cápside/fisiología , Fluorescencia , Virus de la Hepatitis B/fisiología , Técnicas In Vitro , Cinética , Modelos Biológicos
12.
J Mol Biol ; 303(2): 197-211, 2000 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-11023786

RESUMEN

Rice yellow mottle virus (RYMV) and southern bean mosaic virus, cowpea strain (SCPMV) are members of the Sobemovirus genus of RNA-containing viruses. We used electron cryo-microscopy (cryo-EM) and icosahedral image analysis to examine the native structures of these two viruses at 25 A resolution. Both viruses have a single tightly packed capsid layer with 180 subunits assembled on a T=3 icosahedral lattice. Distinctive crown-like pentamers emanate from the 12 5-fold axes of symmetry. The exterior face of SCPMV displays deep valleys along the 2-fold axes and protrusions at the quasi-3-fold axes. While having a similar topography, the surface of RYMV is comparatively smooth. Two concentric shells of density reside beneath the capsid layer of RYMV and SCPMV, which we interpret as ordered regions of genomic RNA. In the presence of divalent cations, SCPMV particles swell and fracture, whereas the expanded form of RYMV is stable. We previously proposed that the cell-to-cell movement of RYMV in xylem involves chelation of Ca(2+) from pit membranes of infected cells, thereby stabilizing the capsid shells and allowing a pathway for spread of RYMV through destabilized membranes. In the context of this model, we propose that the expanded form of RYMV is an intermediate in the in vivo assembly of virions.


Asunto(s)
Microscopía por Crioelectrón , Procesamiento de Imagen Asistido por Computador , Virus de Plantas/química , Virus de Plantas/ultraestructura , Virus ARN/química , Virus ARN/ultraestructura , Secuencia de Aminoácidos , Calcio/metabolismo , Calcio/farmacología , Cápside/química , Cápside/efectos de los fármacos , Cápside/ultraestructura , Cationes Bivalentes/metabolismo , Cationes Bivalentes/farmacología , Cristalografía por Rayos X , Fabaceae/virología , Genoma Viral , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Oryza/virología , Virus de Plantas/efectos de los fármacos , Virus de Plantas/genética , Plantas Medicinales , Virus ARN/efectos de los fármacos , Virus ARN/genética , ARN Viral/genética , ARN Viral/metabolismo , Alineación de Secuencia , Ensamble de Virus/efectos de los fármacos
13.
J Mol Biol ; 273(2): 456-66, 1997 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-9344752

RESUMEN

The different partially folded states of the capsid protein that appear in the disassembly pathway of cowpea severe mosaic virus (CPSMV) were investigated by examining the effects of hydrostatic pressure, sub-zero temperatures and urea. The conformational states of the coat protein were analyzed by their intrinsic fluorescence, binding of bis(8-anilinonaphthalene-1-sulfonate) (bis-ANS) and susceptibility to trypsin digestion. CPSMV could be disassembled by pressure at 2.5 kbar. Intrinsic fluorescence and hydrodynamic measurements showed that pressure-induced dissociation was completely reversible. Virus pressurization in the presence of ribonuclease revealed that viral RNA was not exposed, since it was not digested by the enzyme, suggesting the maintenance of protein-nucleic acid interactions under pressure. When the temperature was decreased to -10 degrees C under pressure, CPSMV disassembly became an irreversible process and in this condition, viral RNA was completely digested by ribonuclease. These results suggest a relationship between protein-RNA interactions and CPSMV assembly. Bis-ANS binding and trypsin digestion of coat proteins revealed that they assume a different conformation when they are denatured by low temperatures under pressure or than when they are denatured by urea at atmospheric pressure. The results indicate that the coat proteins can exist in at least four states: (1) The native conformation in the virus capsid; (2) bound to RNA when the virus is dissociated by pressure at room temperature, assuming a conformation that retains the information for reassembly; (3) free subunits in a molten-globule conformation when the virus is dissociated by low temperature under pressure; and (4) free subunits completely unfolded by high concentrations of urea.


Asunto(s)
Cápside/química , Comovirus/química , Pliegue de Proteína , Proteínas de Unión al ARN/química , Naftalenosulfonatos de Anilina , Cápside/efectos de los fármacos , Frío , Fabaceae/virología , Presión Hidrostática , Modelos Químicos , Plantas Medicinales , Conformación Proteica , ARN Viral/química , Proteínas de Unión al ARN/efectos de los fármacos , Espectrometría de Fluorescencia , Urea/farmacología
14.
J Virol Methods ; 46(2): 255-61, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8188819

RESUMEN

The coat protein from purified particles of pea seedborne mosaic potyvirus (PSbMV) moves in SDS-PAGE with an apparent molecular weight (M(r)) of 36 kDa. However, extracts of PSbMV infected plants prepared with SDS or urea contain PSbMV immunoreactive proteins with apparent M(r) 39 kDa as well as 36 kDa. The low mobility form may be generated from the apparent M(r) 36 kDa form by incubating purified PSbMV particles with healthy plant sap in the presence of denaturing agents. A similar effect is observed with bean yellow mosaic potyvirus, but not with three viruses outside the potyvirus group. Experiments suggest that a soluble plant enzyme is responsible for the conversion, which apparently takes place only in vitro under denaturing conditions. This phenomenon may lead to erroneous conclusions about the M(r) of some viral coat proteins. However, the conversion can be prevented by heat treatment of the plant tissue prior to extraction.


Asunto(s)
Cápside/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida , Fabaceae/microbiología , Plantas Medicinales , Potyvirus/aislamiento & purificación , Cápside/efectos de los fármacos , Dodecil Sulfato de Sodio/farmacología , Urea/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA