Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(14): 3720-3725, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28320935

RESUMEN

Viruses and their hosts can reach balanced states of evolution ensuring mutual survival, which makes it difficult to appreciate the underlying dynamics. To uncover hidden interactions, virus mutants that have lost defense genes may be used. Deletion of the gene that encodes serine protease inhibitor 1 (SPI-1) of rabbitpox virus and vaccinia virus, two closely related orthopoxviruses, prevents their efficient replication in human cells, whereas certain other mammalian cells remain fully permissive. Our high-throughput genome-wide siRNA screen identified host factors that prevent reproduction and spread of the mutant viruses in human cells. More than 20,000 genes were interrogated with individual siRNAs and those that prominently increased replication of the SPI-1 deletion mutant were subjected to a secondary screen. The top hits based on the combined data-replication factor C3 (RFC3), FAM111A, and interferon regulatory factor 2 (IRF2)-were confirmed by custom assays. The siRNAs to RFC1, RFC2, RFC4, and RFC5 mRNAs also enhanced spread of the mutant virus, strengthening the biological significance of the RFC complex as a host restriction factor for poxviruses. Whereas association with proliferating cell nuclear antigen and participation in processive genome replication are common features of FAM111A and RFC, IRF2 is a transcriptional regulator. Microarray analysis, quantitative RT-PCR, and immunoblotting revealed that IRF2 regulated the basal level expression of FAM111A, suggesting that the enhancing effect of depleting IRF2 on replication of the SPI-1 mutant was indirect. Thus, the viral SPI-1 protein and the host IRF2, FAM111A, and RFC complex likely form an interaction network that influences the ability of poxviruses to replicate in human cells.


Asunto(s)
Factor 2 Regulador del Interferón/metabolismo , Orthopoxvirus/fisiología , Receptores Virales/metabolismo , Proteína de Replicación C/metabolismo , Serpinas/genética , Células A549 , Humanos , Análisis por Micromatrices , Mutación , Orthopoxvirus/enzimología , Orthopoxvirus/genética , Infecciones por Poxviridae/metabolismo , Infecciones por Poxviridae/virología , Proteínas Virales/genética , Replicación Viral
2.
J Antimicrob Chemother ; 70(5): 1367-80, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25630650

RESUMEN

OBJECTIVES: ST-246 is one of the key antivirals being developed to fight orthopoxvirus (OPV) infections. Its exact mode of action is not completely understood, but it has been reported to interfere with the wrapping of infectious virions, for which F13L (peripheral membrane protein) and B5R (type I glycoprotein) are required. Here we monitored the appearance of ST-246 resistance to identify its molecular target. METHODS: Vaccinia virus (VACV), cowpox virus (CPXV) and camelpox virus (CMLV) with reduced susceptibility to ST-246 were selected in cell culture and further characterized by antiviral assays and immunofluorescence. A panel of recombinant OPVs was engineered and a putative 3D model of F13L coupled with molecular docking was used to visualize drug-target interaction. The F13L gene of 65 CPXVs was sequenced to investigate F13L amino acid heterogeneity. RESULTS: Amino acid substitutions or insertions were found in the F13L gene of six drug-resistant OPVs and production of four F13L-recombinant viruses confirmed their role(s) in the occurrence of ST-246 resistance. F13L, but not B5R, knockout OPVs showed resistance to ST-246. ST-246 treatment of WT OPVs delocalized F13L- and B5R-encoded proteins and blocked virus wrapping. Putative modelling of F13L and ST-246 revealed a probable pocket into which ST-246 penetrates. None of the identified amino acid changes occurred naturally among newly sequenced or NCBI-derived OPV F13L sequences. CONCLUSIONS: Besides demonstrating that F13L is a direct target of ST-246, we also identified novel F13L residues involved in the interaction with ST-246. These findings are important for ST-246 use in the clinic and crucial for future drug-resistance surveillance programmes.


Asunto(s)
Antivirales/metabolismo , Benzamidas/metabolismo , Virus de la Viruela Vacuna/fisiología , Isoindoles/metabolismo , Orthopoxvirus/fisiología , Fosfolipasas/antagonistas & inhibidores , Virus Vaccinia/fisiología , Ensamble de Virus/efectos de los fármacos , Animales , Virus de la Viruela Vacuna/efectos de los fármacos , Virus de la Viruela Vacuna/enzimología , Virus de la Viruela Vacuna/genética , Farmacorresistencia Viral , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Simulación del Acoplamiento Molecular , Mutación , Orthopoxvirus/efectos de los fármacos , Orthopoxvirus/enzimología , Orthopoxvirus/genética , Fosfolipasas/química , Fosfolipasas/genética , Unión Proteica , Conformación Proteica , Pase Seriado , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/enzimología , Virus Vaccinia/genética , Ensayo de Placa Viral , Cultivo de Virus
3.
J Virol ; 86(13): 7310-25, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22532673

RESUMEN

Cidofovir or (S)-HPMPC is one of the three antiviral drugs that might be used for the treatment of orthopoxvirus infections. (S)-HPMPC and its 2,6-diaminopurine counterpart, (S)-HPMPDAP, have been described to select, in vitro, for drug resistance mutations in the viral DNA polymerase (E9L) gene of vaccinia virus (VACV). Here, to extend our knowledge of drug resistance development among orthopoxviruses, we selected, in vitro, camelpox viruses (CMLV) resistant to (S)-HPMPDAP and identified a single amino acid change, T831I, and a double mutation, A314V+A684V, within E9L. The production of recombinant CMLV and VACV carrying these amino acid substitutions (T831I, A314V, or A314V+A684V) demonstrated clearly their involvement in conferring reduced sensitivity to viral DNA polymerase inhibitors, including (S)-HPMPDAP. Both CMLV and VACV harboring the A314V change showed comparable drug-susceptibility profiles to various antivirals and similar impairments in viral growth. In contrast, the single change T831I and the double change A314V+A684V in VACV were responsible for increased levels of drug resistance and for cross-resistance to viral DNA polymerase antivirals that were not observed with their CMLV counterparts. Each amino acid change accounted for an attenuated phenotype of VACV in vivo. Modeling of E9L suggested that the T→I change at position 831 might abolish hydrogen bonds between E9L and the DNA backbone and have a direct impact on the incorporation of the acyclic nucleoside phosphonates. Our findings demonstrate that drug-resistance development in two related orthopoxvirus species may impact drug-susceptibility profiles and viral fitness differently.


Asunto(s)
Antivirales/farmacología , ADN Polimerasa Dirigida por ADN/genética , Farmacorresistencia Viral , Mutación Missense , Orthopoxvirus/efectos de los fármacos , Virus Vaccinia/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Línea Celular , Cidofovir , Citosina/análogos & derivados , Citosina/farmacología , ADN Polimerasa Dirigida por ADN/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Organofosfonatos/farmacología , Orthopoxvirus/enzimología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Selección Genética , Virus Vaccinia/enzimología , Ensayo de Placa Viral
4.
Virus Genes ; 41(3): 309-18, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20703935

RESUMEN

Protein modification by ubiquitin or ubiquitin-like polypeptides is important for the fate and functions of the majority of proteins in the eukaryotic cell and can be involved in regulation of various biological processes, including protein metabolism (degradation), protein transport to several cellular compartments, rearrangement of cytoskeleton, and transcription of cytoprotective genes. The accumulated experimental data suggest that the ankyrin-F-box-like and BTB-kelch-like proteins of orthopoxviruses, represented by the largest viral multigene families, interact with the cellular Cullin-1- and Cullin-3-containing ubiquitin-protein ligases, respectively. In addition, orthopoxviruses code for their own RING-domain-containing ubiquitin ligase. In this review, this author discusses the differences between variola (smallpox), monkeypox, cowpox, vaccinia, and ectromelia (mousepox) viruses in the organization of ankyrin-F-box and BTB-kelch protein families and their likely functions.


Asunto(s)
Orthopoxvirus/metabolismo , Infecciones por Poxviridae/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Orthopoxvirus/química , Orthopoxvirus/enzimología , Orthopoxvirus/genética , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/virología , Unión Proteica , Ubiquitina-Proteína Ligasas/genética , Proteínas Virales/química , Proteínas Virales/genética
5.
J Virol ; 83(5): 2099-108, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19109393

RESUMEN

Cellular homeostasis depends on an intricate balance of protein expression and degradation. The ubiquitin-proteasome pathway plays a crucial role in specifically targeting proteins tagged with ubiquitin for destruction. This degradation can be effectively blocked by both chemically synthesized and natural proteasome inhibitors. Poxviruses encode a number of proteins that exploit the ubiquitin-proteasome system, including virally encoded ubiquitin molecules and ubiquitin ligases, as well as BTB/kelch proteins and F-box proteins, which interact with cellular ubiquitin ligases. Here we show that poxvirus infection was dramatically affected by a range of proteasome inhibitors, including MG132, MG115, lactacystin, and bortezomib (Velcade). Confocal microscopy demonstrated that infected cells treated with MG132 or bortezomib lacked viral replication factories within the cytoplasm. This was accompanied by the absence of late gene expression and DNA replication; however, early gene expression occurred unabated. Proteasomal inhibition with MG132 or bortezomib also had dramatic effects on viral titers, severely blocking viral replication and propagation. The effects of MG132 on poxvirus infection were reversible upon washout, resulting in the production of late genes and viral replication factories. Significantly, the addition of an ubiquitin-activating enzyme (E1) inhibitor had a similar affect on late and early protein expression. Together, our data suggests that a functional ubiquitin-proteasome system is required during poxvirus infection.


Asunto(s)
Orthopoxvirus/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Chlorocebus aethiops , Cricetinae , Replicación del ADN/efectos de los fármacos , ADN Viral/efectos de los fármacos , Células HeLa , Humanos , Ratones , Orthopoxvirus/genética , Orthopoxvirus/fisiología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/metabolismo , Proteínas Virales/antagonistas & inhibidores
6.
Virol J ; 4: 8, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17224068

RESUMEN

The potential use of variola virus, the causative agent of smallpox, as a bioweapon and the endemic presence of monkeypox virus in Africa demonstrate the need for better therapies for orthopoxvirus infections. Chemotherapeutic approaches to control viral infections have been less successful than those targeting bacterial infections. While bacteria commonly reproduce themselves outside of cells and have metabolic functions against which antibiotics can be directed, viruses replicate in the host cells using the cells' metabolic pathways. This makes it very difficult to selectively target the virus without damaging the host. Therefore, the development of antiviral drugs against poxviruses has initially focused on unique properties of the viral replication cycle or of viral proteins that can be selectively targeted. However, recent advances in molecular biology have provided insights into host factors that represent novel drug targets. The latest anti-poxvirus drugs are kinase inhibitors, which were originally developed to treat cancer progression but in addition block egress of poxviruses from infected cells. This review will summarize the current understanding of anti-poxvirus drugs and will give an overview of the development of the latest second generation poxvirus drugs.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Orthopoxvirus/efectos de los fármacos , Fosfotransferasas/antagonistas & inhibidores , Animales , Antivirales/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratones , Orthopoxvirus/clasificación , Orthopoxvirus/enzimología , Orthopoxvirus/fisiología , Infecciones por Poxviridae/tratamiento farmacológico , Infecciones por Poxviridae/virología , Proteínas Virales/efectos de los fármacos , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
7.
J Mol Biol ; 354(3): 529-35, 2005 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-16253270

RESUMEN

A putative RNase P RNA gene in camelpox virus, one of the orthopoxviruses, was cloned and transcribed in vitro. No RNase P activity could be detected in vitro from camelpox virus RNase P RNA alone, or by addition of the Escherichia coli RNase P protein subunit to reaction mixtures. Camelpox virus RNase P RNA reconstituted in vitro with camel or HeLa cell extracts, which were pre-treated with micrococcal nuclease to degrade the endogenous RNase P RNA, showed no RNase P activity. Vaccinia virus, another orthopoxvirus, showed no RNase P activity in vaccinia-infected HeLa cells, even though transcription of the vaccinia RNase P RNA could be identified in the cells by both Northern blot and RNase protection assay. Camelpox virus RNase P RNA inhibited an endogenous HeLa RNase P activity by 20% in our assays. The 5S RNA showed no significant inhibition in this assay.


Asunto(s)
Orthopoxvirus/enzimología , Orthopoxvirus/genética , ARN Viral/química , ARN Viral/metabolismo , Ribonucleasa P/genética , Animales , Secuencia de Bases , Camelus , Escherichia coli/enzimología , Células HeLa , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Ribosómico 5S/genética , ARN Ribosómico 5S/metabolismo , ARN Viral/genética , Virus Vaccinia/genética
8.
Virus Genes ; 9(3): 231-45, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7597802

RESUMEN

The genome nucleotide sequences of two strains of variola major virus and one strain of vaccinia virus were compared. One hundred and sixty-eight short (less than 100 bp in length) and eight long (more than 900 bp in length) deletions, four deletion/insertion regions, and four regions of multiple mutational differences between variola and vaccinia virus DNAs were revealed. Short deletions generally occur at directly repeated sequences of 3-21 bp. Long deletions showed no evidence of repeated sequences at their points of junction. We suggest the presence of a consensus sequence characteristic of these junctions and propose that there is a virus-encoded enzyme that produces this nonhomologous recombination/deletion in the cytoplasm of the infected cell.


Asunto(s)
ADN Viral/genética , Genoma Viral , Orthopoxvirus/genética , Eliminación de Secuencia , Secuencia de Bases , Secuencia de Consenso , Análisis Mutacional de ADN , Datos de Secuencia Molecular , Orthopoxvirus/enzimología , Recombinación Genética , Secuencias Repetitivas de Ácidos Nucleicos , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Virus Vaccinia/enzimología , Virus Vaccinia/genética , Virus de la Viruela/enzimología , Virus de la Viruela/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...