Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Virol ; 94(22)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32907985

RESUMEN

Mx proteins are interferon (IFN) type I (α/ß)- and type III (λ)-induced effector proteins with intrinsic antiviral activity. Mammalian Mx proteins show different subcellular localizations and distinct yet partially overlapping viral specificities. However, the precise mechanism(s) of antiviral action are still unresolved. Human MxA accumulates in the cytoplasm and inhibits a wide variety of RNA and DNA viruses, among them influenza A virus (IAV). In contrast, MxB, the second human Mx protein, localizes via its amino (N) terminus to the outer nuclear membrane at or near nuclear pores and inhibits the nuclear import of incoming human immunodeficiency viruses (HIV) and herpesviruses, but not that of IAV. Here, we evaluated whether the antiviral specificity of MxB is determined by its subcellular localization. For this purpose, we redirected MxB to the nucleus or cytoplasm by either attaching a nuclear localization signal to its N terminus or by exchanging the N terminus of MxB with that of MxA. Interestingly, ectopic expression of these MxB variants in the nucleus or in the cytoplasm rendered the host cells resistant to IAV, revealing that the capacity of MxB to block IAV replication critically depends on the site where the protein accumulates in the infected cell. Furthermore, coimmunoprecipitation (co-IP) assays demonstrated that MxB physically interacted with the nucleoprotein (NP) of IAV. Taken together, the data indicate that the subcellular localization of the MxB protein plays a pivotal role in determining its antiviral specificity.IMPORTANCE The interferon system plays a pivotal role in the defense against viral infections. The dynamin-related Mx proteins form a small family of interferon-induced effector proteins with distinct antiviral specificities and subcellular localizations. So far, it is not clear whether the different virus specificities of Mx proteins are the result of distinct mechanisms of action or are due rather to their different subcellular localization. We show here that the human MxB protein, normally localized to the outer membrane of the cell nucleus, acquires antiviral activity against IAV when redirected to the nucleus or cytoplasm, subcellular sites where other members of the Mx protein family efficiently interfere with IAV replication. Our findings thus strongly suggest that Mx proteins act primarily through a common mechanism and that their viral specificity is at least in part determined by their individual subcellular localization.


Asunto(s)
Antivirales/metabolismo , Virus de la Influenza A/efectos de los fármacos , Proteínas de Resistencia a Mixovirus/metabolismo , Transporte Activo de Núcleo Celular , Antivirales/farmacología , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Virus de la Influenza A/metabolismo , Interferones/metabolismo , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/farmacología , Nucleoproteínas , Replicación Viral/efectos de los fármacos
2.
Acta biol. colomb ; 24(3): 493-502, Sep.-Dec. 2019. graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1054643

RESUMEN

ABSTRACT Hepatitis C Virus belongs to the Flaviviridae family. One proposed mechanism of HCV persistence in the ability to infect hematopoietic cells, including Dendritic cells (DCs). HCV infection of DCs could impair their functions that represent one of the mechanisms, thus hampering viral clearance by the host immune system. Among HCV-encoded proteins, the highly conserved Core protein has been suggested to be responsible for the immunomodulatory properties of this Hepacivirus. Recombinant viral vectors expressing the HCV Core protein and allowing its transduction and therefore the expression of the protein into DCs could be useful tools for the analysis of the properties of the Core protein. Vaccinia Virus and retrovirus have been used to transduce human DCs. Likewise, gene transfer into DCs using Semliki Forest Virus has been reported. This study aimed to express the HCV Core protein in human monocyte-derived DCs using an SFV vector, in which the subgenomic RNA encoding the structural proteins was replaced by the HCV Core sequence and then analyze the effects of its expression on DCs functions.


RESUMEN El virus de la Hepatitis C (VHC) pertenece a la familia Flaviviridae. Uno de los mecanismos propuestos de la persistencia del VHC es la capacidad de infectar células hematopoyéticas, incluidas las células dendríticas (DCs). La infección por VHC de DCs podría alterar sus funciones y corresponde a uno de los mecanismos que impiden el aclaramiento de la infección por VHC por el sistema inmunitario del hospedero. Entre las proteínas codificadas por el VHC, se ha sugerido que la proteína Core, altamente conservada, es responsable de las propiedades inmunomoduladoras de este Hepacivirus. Los vectores virales recombinantes que expresan la proteína Core y permiten su transducción a DCs podrían ser herramientas útiles para el análisis de las propiedades de esta proteína. El virus Vaccinia y el retrovirus se han utilizado para la transducción de DCs humanas. Del mismo modo, la transducción de DCs usando el virus del bosque de Semliki ha sido reportada. El objetivo de este estudio fue expresar la proteína Core de VHC en DCs derivadas de monocitos humanos utilizando un vector de SFV, en el que el ARN subgenómico que codifica las proteínas estructurales fue reemplazado por la secuencia Core del VHC y evaluar los efectos de su expresión en las funciones de DCs.

3.
Annu Rev Virol ; 5(1): 33-51, 2018 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-29958082

RESUMEN

The discovery of the Mx gene-dependent, innate resistance of mice against influenza virus was a matter of pure chance. Although the subsequent analysis of this antiviral resistance was guided by straightforward logic, it nevertheless led us into many blind alleys and was full of surprising turns and twists. Unexpectedly, this research resulted in the identification of one of the first interferon-stimulated genes and provided a new view of interferon action. It also showed that in many species, MX proteins have activities against a broad range of viruses. To this day, Mx research continues to flourish and to provide insights into the never-ending battle between viruses and their hosts.


Asunto(s)
Investigación Biomédica/historia , Resistencia a la Enfermedad , Inmunidad Innata , Proteínas de Resistencia a Mixovirus/metabolismo , Virus/inmunología , Animales , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Interferones/metabolismo
4.
Nat Commun ; 9(1): 1980, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773792

RESUMEN

The type I interferon (IFN) system plays an important role in controlling herpesvirus infections, but it is unclear which IFN-mediated effectors interfere with herpesvirus replication. Here we report that human myxovirus resistance protein B (MxB, also designated Mx2) is a potent human herpesvirus restriction factor in the context of IFN. We demonstrate that ectopic MxB expression restricts a range of herpesviruses from the Alphaherpesvirinae and Gammaherpesvirinae, including herpes simplex virus 1 and 2 (HSV-1 and HSV-2), and Kaposi's sarcoma-associated herpesvirus (KSHV). MxB restriction of HSV-1 and HSV-2 requires GTPase function, in contrast to restriction of lentiviruses. MxB inhibits the delivery of incoming HSV-1 DNA to the nucleus and the appearance of empty capsids, but not the capsid delivery to the cytoplasm or tegument dissociation from the capsid. Our study identifies MxB as a potent pan-herpesvirus restriction factor which blocks the uncoating of viral DNA from the incoming viral capsid.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Herpesviridae/fisiología , Interferón Tipo I/inmunología , Proteínas de Resistencia a Mixovirus/inmunología , Replicación Viral/inmunología , Cápside/inmunología , Proteínas de la Cápside/inmunología , Línea Celular Tumoral , Núcleo Celular/inmunología , Núcleo Celular/virología , Citoplasma , ADN Viral/inmunología , Células HEK293 , Herpesviridae/patogenicidad , Infecciones por Herpesviridae/virología , Humanos , Proteínas de Resistencia a Mixovirus/genética , ARN Interferente Pequeño/metabolismo , Desencapsidación Viral/inmunología
5.
J Vis Exp ; (116)2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27842357

RESUMEN

The formation of oligomeric complexes is a crucial prerequisite for the proper structure and function of many proteins. The interferon-induced antiviral effector protein MxA exerts a broad antiviral activity against many viruses. MxA is a dynamin-like GTPase and has the capacity to form oligomeric structures of higher order. However, whether oligomerization of MxA is required for its antiviral activity is an issue of debate. We describe here a simple protocol to assess the oligomeric state of endogenously or ectopically expressed MxA in the cytoplasmic fraction of human cell lines by non-denaturing polyacrylamide gel electrophoresis (PAGE) in combination with Western blot analysis. A critical step of the protocol is the choice of detergents to prevent aggregation and/or precipitation of proteins particularly associated with cellular membranes such as MxA, without interfering with its enzymatic activity. Another crucial aspect of the protocol is the irreversible protection of the free thiol groups of cysteine residues by iodoacetamide to prevent artificial interactions of the protein. This protocol is suitable for a simple assessment of the oligomeric state of MxA and furthermore allows a direct correlation of the antiviral activity of MxA interface mutants with their respective oligomeric states.


Asunto(s)
Electroforesis en Gel de Poliacrilamida , Proteínas de Resistencia a Mixovirus , Antivirales , Western Blotting , Electroforesis , GTP Fosfohidrolasas , Interacciones Huésped-Patógeno , Humanos , Interferones , Dominios y Motivos de Interacción de Proteínas
6.
Retrovirology ; 13(1): 62, 2016 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-27595568

RESUMEN

BACKGROUND: Mucosal HIV-1 transmission predominantly results in a single transmitted/founder (T/F) virus establishing infection in the new host despite the generally high genetic diversity of the transmitter virus population. To what extent HIV-1 transmission is a stochastic process or driven by selective forces that allow T/F viruses best to overcome bottlenecks in transmission has not been conclusively resolved. Building on prior investigations that suggest HIV-1 envelope (Env) features to contribute in the selection process during transmission, we compared phenotypic virus characteristics of nine HIV-1 subtype B transmission pairs, six men who have sex with men and three male-to-female transmission pairs. RESULTS: All recipients were identified early in acute infection and harbored based on extensive sequencing analysis a single T/F virus allowing a controlled analysis of virus properties in matched transmission pairs. Recipient and transmitter viruses from the closest time point to transmission showed no signs of selection for specific Env modifications such as variable loop length and glycosylation. Recipient viruses were resistant to circulating plasma antibodies of the transmitter and also showed no altered sensitivity to a large panel of entry inhibitors and neutralizing antibodies. The recipient virus did not consistently differ from the transmitter virus in terms of entry kinetics, cell-cell transmission and replicative capacity in primary cells. Our paired analysis revealed a higher sensitivity of several recipient virus isolates to interferon-α (IFNα) which suggests that resistance to IFNα cannot be a general driving force in T/F establishment. CONCLUSIONS: With the exception of increased IFNα sensitivity, none of the phenotypic virus properties we investigated clearly distinguished T/F viruses from their matched transmitter viruses supporting the notion that at least in subtype B infection HIV-1 transmission is to a considerable extent stochastic.


Asunto(s)
Infecciones por VIH/transmisión , VIH-1/genética , VIH-1/fisiología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Enfermedad Aguda , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Femenino , Variación Genética , Infecciones por VIH/virología , VIH-1/aislamiento & purificación , Homosexualidad Masculina , Humanos , Interferón-alfa/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Masculino , Pruebas de Neutralización , Fenotipo , Análisis de Secuencia de ADN , Procesos Estocásticos , Tropismo Viral , Internalización del Virus , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
7.
J Biol Chem ; 290(50): 29893-906, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26507657

RESUMEN

The IFN-induced human myxovirus resistance protein A (MxA) exhibits a broad antiviral activity against many viruses, including influenza A virus (IAV). MxA belongs to the family of dynamin-like GTPases and assembles in vitro into dimers, tetramers, and oligomeric ring-like structures. The molecular mechanism of action remains to be elucidated. Furthermore, it is not clear whether MxA exerts its antiviral activity in a monomeric and/or multimeric form. Using a set of MxA mutants that form complexes with defined stoichiometry, we observed that, in the presence of guanosine 5'-O-(thiotriphosphate), purified MxA disassembled into tetramers and dimers. Dimeric forms did not further disassemble into monomers. Infection experiments revealed that besides wild-type MxA, dimeric and monomeric variants of MxA also efficiently restricted IAV at a replication step after primary transcription. Moreover, only dimeric MxA was able to form stable complexes with the nucleoprotein (NP) of IAV. MxA interacted with NP independently of other viral components. Interestingly, the dimeric form of MxA was able to efficiently bind to NP from several MxA-sensitive strains but interacted much more weakly with NP from the MxA-resistant PR8 strain derived from the H1N1 1918 lineage. Taken together, these data suggest that, during infection, a fraction of MxA disassembles into dimers that bind to NP synthesized following primary transcription in the cytoplasm, thereby preventing viral replication.


Asunto(s)
Biopolímeros/metabolismo , Guanosina Trifosfato/metabolismo , Virus de la Influenza A/metabolismo , Proteínas de Resistencia a Mixovirus/metabolismo , Animales , Línea Celular , Humanos
8.
Retrovirology ; 9: 27, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22458358

RESUMEN

BACKGROUND: The various classes of small noncoding RNAs (sncRNAs) are important regulators of gene expression across divergent types of organisms. While a rapidly increasing number of sncRNAs has been identified over recent years, the isolation of sncRNAs of low abundance remains challenging. Virally encoded sncRNAs, particularly those of RNA viruses, can be expressed at very low levels. This is best illustrated by HIV-1 where virus encoded sncRNAs represent approximately 0.1-1.0% of all sncRNAs in HIV-1 infected cells or were found to be undetected. Thus, we applied a novel, sequence targeted enrichment strategy to capture HIV-1 derived sncRNAs in HIV-1 infected primary CD4+ T-lymphocytes and macrophages that allows a greater than 100-fold enrichment of low abundant sncRNAs. RESULTS: Eight hundred and ninety-two individual HIV-1 sncRNAs were cloned and sequenced from nine different sncRNA libraries derived from five independent experiments. These clones represent up to 90% of all sncRNA clones in the generated libraries. Two hundred and sixteen HIV-1 sncRNAs were distinguishable as unique clones. They are spread throughout the HIV-1 genome, however, forming certain clusters, and almost 10% show an antisense orientation. The length of HIV-1 sncRNAs varies between 16 and 89 nucleotides with an unexpected peak at 31 to 50 nucleotides, thus, longer than cellular microRNAs or short-interfering RNAs (siRNAs). Exemplary HIV-1 sncRNAs were also generated in cells infected with different primary HIV-1 isolates and can inhibit HIV-1 replication. CONCLUSIONS: HIV-1 infected cells generate virally encoded sncRNAs, which might play a role in the HIV-1 life cycle. Furthermore, the enormous capacity to enrich low abundance sncRNAs in a sequence specific manner highly recommends our selection strategy for any type of investigation where origin or target sequences of the sought-after sncRNAs are known.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Regulación Viral de la Expresión Génica , VIH-1/patogenicidad , Macrófagos/virología , ARN Pequeño no Traducido/metabolismo , Células Cultivadas , VIH-1/genética , Humanos , ARN Pequeño no Traducido/genética , ARN Viral/genética , ARN Viral/metabolismo
9.
J Biol Chem ; 286(40): 34743-51, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21859714

RESUMEN

Mx proteins are a family of large GTPases that are induced exclusively by interferon-α/ß and have a broad antiviral activity against several viruses, including influenza A virus (IAV). Although the antiviral activities of mouse Mx1 and human MxA have been studied extensively, the molecular mechanism of action remains largely unsolved. Because no direct interaction between Mx proteins and IAV proteins or RNA had been demonstrated so far, we addressed the question of whether Mx protein would interact with cellular proteins required for efficient replication of IAV. Immunoprecipitation of MxA revealed its association with two closely related RNA helicases, UAP56 and URH49. UAP56 and its paralog URH49 play an important role in IAV replication and are involved in nuclear export of IAV mRNAs and prevention of dsRNA accumulation in infected cells. In vitro binding assays with purified recombinant proteins revealed that MxA formed a direct complex with the RNA helicases. In addition, recombinant mouse Mx1 was also able to bind to UAP56 or URH49. Furthermore, the complex formation between cytoplasmic MxA and UAP56 or URH49 occurred in the perinuclear region, whereas nuclear Mx1 interacted with UAP56 or URH49 in distinct dots in the nucleus. Taken together, our data reveal that Mx proteins exerting antiviral activity can directly bind to the two cellular DExD/H box RNA helicases UAP56 and URH49. Moreover, the observed subcellular localization of the Mx-RNA helicase complexes coincides with the subcellular localization, where human MxA and mouse Mx1 proteins act antivirally. On the basis of these data, we propose that Mx proteins exert their antiviral activity against IAV by interfering with the function of the RNA helicases UAP56 and URH49.


Asunto(s)
Antivirales/metabolismo , ARN Helicasas DEAD-box/metabolismo , Proteínas de Unión al GTP/metabolismo , Interferones/metabolismo , ARN Helicasas/metabolismo , Células 3T3 , Animales , Antivirales/farmacología , Citoplasma/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Virus de la Influenza A/metabolismo , Ratones , Proteínas de Resistencia a Mixovirus , Unión Proteica , ARN Helicasas/química
10.
J Virol ; 85(17): 8646-55, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21680511

RESUMEN

The cellular DEAD box RNA helicase UAP56 plays a pivotal role in the efficient transcription/replication of influenza A virus. UAP56 is recruited by the nucleoprotein (NP) of influenza A viruses, and recent data revealed that the RNA helicase is required for the nuclear export of a subset of spliced and unspliced viral mRNAs. The fact that influenza viruses do not produce detectable amounts of double-stranded RNA (dsRNA) intermediates during transcription/replication suggests the involvement of cellular RNA helicases. Hence, we examined whether the RNA-unwinding activity of UAP56 or its paralog URH49 plays a role in preventing the accumulation of dsRNA during infection. First, our data showed that not only UAP56 but also its paralog URH49 can interact with NPs of avian and human influenza A viruses. The small interfering RNA (siRNA)-mediated depletion of either RNA helicase reduced the transport of M1 and hemagglutinin (HA) mRNAs and, to a lesser extent, NP and NS1 mRNAs into the cytoplasm. Moreover, we found that virus infection of UAP56-depleted cells leads to the rapid accumulation of dsRNA in the perinuclear region. In parallel, we observed a robust virus-mediated activation of dsRNA-dependent protein kinase R (PKR), indicating that the cellular RNA helicase UAP56 may be recruited by influenza virus to prevent dsRNA formation. The accumulation of dsRNA was blocked when actinomycin D or cycloheximide was used to inhibit viral transcription/replication or translation, respectively. In summary, we demonstrate that UAP56 is utilized by influenza A viruses to prevent the formation of dsRNA and, hence, the activation of the innate immune response.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Replicación Viral , Humanos , Proteínas de la Nucleocápside , Unión Proteica , Proteínas de Unión al ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo
11.
Arch Virol ; 154(1): 109-14, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19034603

RESUMEN

Influenza A virus causes prevalent respiratory tract infections in humans. Small interfering RNA (siRNA) and antisense oligonucleotides (asODNs) have been used previously for silencing the RNA genome of influenza virus. Here, we explored the use of partially double-stranded oligodeoxynucleotides (dsODNs) to suppress the production of influenza A virus in cell cultures and animal models. We were able to inhibit influenza A virus replication in cultured human lung cells as well as in the lungs of infected C57BL/6 mice by treatment with dsODN 3-h post-infection. In about 20% of the cases (15/77) the titer was reduced by 10- to 100-fold and in 10% up to 1,000-fold. The antiviral effects of dsODNs were dose-dependent, sequence-dependent and comparable to those of its antisense and siRNA analogues. Thus, dsODNs may be developed as an additional class of nucleic acids for the inhibition of influenza virus replication.


Asunto(s)
ADN/farmacología , Virus de la Influenza A/efectos de los fármacos , Oligodesoxirribonucleótidos/farmacología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , ADN/genética , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/química , Células Vero
12.
FEBS Lett ; 581(28): 5349-54, 2007 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-17967423

RESUMEN

AF6 and its rat homologue afadin are multidomain proteins localized at cell junctions and involved in intercellular adhesion. AF6 interacts via its PDZ domain with nectin-1 at epithelial adherens junctions. Nectin-1 serves as a mediator of cell-to-cell spread for Herpes simplex virus 1 (HSV-1). We analyzed the role of AF6 protein in the viral spread and nectin-1 clustering at cell-cell contacts by knockdown of AF6 in epithelial cells. AF6 knockdown reduced efficiency of HSV-1 spreading, however, the clustering of nectin-1 at cell-cell contacts was not affected. Thus, AF6 protein is important for spreading of HSV-1 in epithelial cells, independently of nectin clustering, possibly by stabilization of the E-cadherin-dependent cell adhesion.


Asunto(s)
Herpesvirus Humano 1/fisiología , Cinesinas/metabolismo , Miosinas/metabolismo , Internalización del Virus , Animales , Moléculas de Adhesión Celular/metabolismo , Comunicación Celular , Línea Celular , Chlorocebus aethiops , Humanos , Cinesinas/genética , Miosinas/genética , Nectinas
13.
Eur J Immunol ; 37(8): 2317-25, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17578844

RESUMEN

IL-12, the critical factor for the generation of the Th1 type immune response, is produced by dendritic cells (DC) upon stimulation with LPS. Different signal pathways mediate LPS-induced expression of IL-12 and involve PI3K, MAPK and the transcription factor NF-kappaB. Here, we show that the kinase Raf is involved in the expression of IL-12 in human DC stimulated by LPS. We demonstrate that Raf regulates the expression of the IL-12 subunit p40 not via the kinase MEK, the major effector of Raf in growth factor-dependent signaling, but via the receptor-interacting protein 2 (RIP2) using specific inhibitors for MAPK pathways. RIP2 is a kinase participating in LPS/Toll-like receptor 4 signaling. Knockdown of RIP2 by siRNA inhibited LPS-dependent expression of IL-12 p40. In addition, knockdown of RIP2 reduced phosphorylation of p38 MAPK, ERK and IkappaBalpha, which are known upstream regulators of IL-12 production. Thus, in human DC LPS stimulates a signal cascade that involves the Raf-dependent activation of RIP2 leading to expression of IL-12 p40.


Asunto(s)
Células Dendríticas/inmunología , Proteínas I-kappa B/inmunología , Subunidad p40 de la Interleucina-12/biosíntesis , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/inmunología , Transducción de Señal/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Western Blotting , Células Cultivadas , Células Dendríticas/metabolismo , Expresión Génica/inmunología , Humanos , Proteínas I-kappa B/metabolismo , Lipopolisacáridos/inmunología , Monocitos/citología , Monocitos/inmunología , Inhibidor NF-kappaB alfa , ARN Interferente Pequeño , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
J Gen Virol ; 88(Pt 7): 1952-1959, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17554028

RESUMEN

Type I interferons (IFNs) are essential components of the innate immune system. This study characterized the distinct IFN sensitivities of two closely related Semliki Forest virus (SFV) strains in cell culture. The virulent L10 strain was derived from the original virus isolate by propagation in mice. In contrast, the avirulent SFV strain, designated V42, was derived from an earlier passage of the original virus isolated from mosquitoes. The virulent L10 strain produced a cytopathic effect (CPE) in IFN-treated cells and the production of infectious virus was only two orders of magnitude lower compared with untreated cells. In contrast, the avirulent V42 exerted no CPE in IFN-treated cells and production of infectious virus was four orders of magnitude lower compared with untreated cells. The reduced CPE in IFN-treated cells infected with the avirulent V42 strain was due to inhibition of productive infection and not to reduced cell death. The virulent L10 strain synthesized less genomic RNA but more non-structural proteins than the avirulent V42 strain, suggesting more efficient translation of the L10 genomic RNA. Using a cell line unable to produce IFN, it was shown that the reduced susceptibility of the L10 strain to the action of IFN was not due to reduced IFN induction. Hence, the reduced susceptibility of the virulent L10 strain to the action of IFN allows it to overcome the established IFN-induced antiviral state of the cell, thereby increasing its virulence.


Asunto(s)
Interferón Tipo I/farmacología , Virus de los Bosques Semliki/efectos de los fármacos , Virus de los Bosques Semliki/patogenicidad , Adaptación Fisiológica , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Efecto Citopatogénico Viral , Farmacorresistencia Viral , Humanos , Ratones , Ratones Noqueados , Células 3T3 NIH , ARN Viral/biosíntesis , ARN Viral/genética , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Proteínas Recombinantes , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/fisiología , Especificidad de la Especie , Células Vero , Virulencia , Replicación Viral
15.
J Gen Virol ; 88(Pt 5): 1403-1409, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17412966

RESUMEN

Non-structural protein NS1 of influenza A virus counteracts the host immune response by blocking the synthesis of type I interferon (IFN). As deletion of the complete NS1 gene has to date been reported only in the human H1N1 strain A/PR/8/34, it remained unclear whether NS1 is a non-essential virulence factor in other influenza A virus strains as well. In this report, the properties of NS1-deficient mutants derived from strain SC35M (H7N7) are described. A mutant of SC35M that completely lacks the NS1 gene was an excellent inducer of IFN in mammalian and avian cells in culture and, consequently, was able to multiply efficiently only in cell lines with defects in the type I IFN system. Virus mutants carrying C-terminally truncated versions of NS1 were less powerful inducers of IFN and were attenuated less strongly in human A549 cells. Although attenuated in wild-type mice, these mutants remained highly pathogenic for mice lacking the IFN-regulated antiviral factor Mx1. In contrast, the NS1-deficient SC35M mutant was completely non-pathogenic for wild-type mice, but remained pathogenic for mice lacking Mx1 and double-stranded RNA-activated protein kinase (PKR). Wild-type SC35M, but not the NS1-deficient mutant virus, was able to replicate in the upper respiratory tract of birds, but neither virus induced severe disease in adult chickens. Altogether, this study supports the view that NS1 represents a non-essential virulence factor of different influenza A viruses.


Asunto(s)
Subtipo H7N7 del Virus de la Influenza A/fisiología , Gripe Aviar/inmunología , Proteínas no Estructurales Virales/deficiencia , Animales , Línea Celular , Pollos , Chlorocebus aethiops , Perros , Humanos , Subtipo H7N7 del Virus de la Influenza A/clasificación , Subtipo H7N7 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana , Interferón beta/genética , Interferones/antagonistas & inhibidores , Pulmón/inmunología , Pulmón/virología , Ratones , Infecciones por Orthomyxoviridae , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero , Virulencia
16.
Vaccine ; 25(16): 2974-83, 2007 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-17303293

RESUMEN

Live attenuated measles virus (MV) vaccines have an impressive record of safety, efficacy and ability to induce life-long immunity against measles infection. Using reverse genetics technology, such negative-strand RNA viruses can now be rescued from cloned DNA. This technology allows the insertion of exogenous genes encoding foreign antigens into the MV genome in such a way that they can be expressed by the MV vaccine strain, without affecting virus structure, propagation and cell targeting. Recombinant viruses rescued from cloned cDNA induce immune responses against both measles virus and the cloned antigens. The tolerability of MV to gene(s) insertion makes it an attractive flexible vector system, especially if broad immune responses are required. The fact that measles replication strictly occurs in the cytoplasm of infected cells without DNA intermediate has important biosafety implications and adds to the attractiveness of MV as a vector. In this article we report the characteristics of reporter gene expression (GFP, LacZ and CAT) and the biochemical, biophysical and immunological properties of recombinant MV expressing heterologous antigens of simian immunogeficiency virus (SIV).


Asunto(s)
Antígenos Virales/metabolismo , Vacuna Antisarampión/inmunología , Virus del Sarampión/inmunología , Sarampión/prevención & control , Vacunas Atenuadas/administración & dosificación , Animales , Antígenos Virales/genética , Línea Celular , Chlorocebus aethiops , Vectores Genéticos , Sarampión/virología , Vacuna Antisarampión/genética , Virus del Sarampión/genética , Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Células Vero
17.
Exp Dermatol ; 15(11): 865-74, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17002683

RESUMEN

Mesenchymal stem cells (MSCs) represent a new tool for delivery of therapeutic agents to tumor cells. In this study, we have evaluated the anti-tumor activity of human MSCs stably transduced with a retroviral vector expressing the cytokine interleukin-12 (IL-12) in a mouse melanoma model. Application of MSC(IL-12) but not control MSCs strongly reduced the formation of lung metastases of B16F10 melanoma cells. The activity of the MSC(IL-12) cells was dependent on the presence of natural killer (NK) cells in this experimental setting. Further, MSC(IL-12) cells elicited a pronounced retardation of tumor growth and led to prolonged survival when injected into established subcutaneous melanoma in a therapeutic regimen. The therapeutic effect of the MSC(IL-12) was in part mediated by CD8(+) T cells, while NK cells and CD4(+) T cells appeared to play a minor role. The anti-tumor effect of MSC(IL-12) cells was of similar efficiency as observed for application of naked plasmid DNA encoding IL-12. The presented data demonstrate that these two different strategies can induce a similar therapeutic anti-tumor efficacy in the mouse melanoma tumor model.


Asunto(s)
Inmunoterapia/métodos , Interleucina-12/genética , Neoplasias Pulmonares/terapia , Melanoma/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Animales , Biopsia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inmunofenotipificación , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-12/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Melanoma/inmunología , Melanoma/secundario , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Ratas , Retroviridae/genética , Bazo/citología , Transducción Genética
18.
Hum Gene Ther ; 17(8): 859-70, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16942445

RESUMEN

The antitumor efficacy of human melanoma-associated antigen (hgp100) and chemokine CCL21 in combination with interleukin-12 (IL-12) was evaluated in a syngeneic melanoma mouse model. The rationale for this approach was based on previous studies showing that the efficacy of IL-12 therapy in melanoma patients correlated with the presence of antibodies against tumor-associated antigens. We have previously shown that application of xenogeneic human gp100 DNA (hgp100 DNA) is protective against mouse B16 melanoma. Furthermore, the chemokine CCL21 has the ability to chemoattract both dendritic cells (DCs) and T lymphocytes. We show here that intratumoral injection of IL-12-encoding DNA (IL-12 DNA) in combination with hgp100- encoding DNA (hgp100 DNA) into tumor-bearing mice led to a strong antitumor effect. Coapplication of IL- 12 DNA with CCL21-encoding DNA (CCL21 DNA) or recombinant CCL21 (recCCL21) protein also showed some efficacy. Triple therapy with IL-12 DNA, hgp100 DNA, and CCL21 DNA, however, showed less effect on tumor growth than double therapy with IL-12 DNA and hgp100 DNA. These findings open a new route of investigation of IL-12 and gp100 or other tumor-associated antigens in the immunotherapy of a variety of tumors.


Asunto(s)
Vacunas contra el Cáncer , Quimiocinas CC , Interleucina-12 , Melanoma/terapia , Glicoproteínas de Membrana , Vacunas de ADN , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Quimiocinas CC/genética , Quimiocinas CC/inmunología , Femenino , Humanos , Interleucina-12/genética , Interleucina-12/inmunología , Melanoma/genética , Melanoma/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Trasplante de Neoplasias/métodos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Trasplante Heterólogo , Trasplante Isogénico , Vacunación , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Antígeno gp100 del Melanoma
19.
Ophthalmic Res ; 37(2): 67-71, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15746561

RESUMEN

BACKGROUND: Ex vivo gene transfer to donor corneas using adenoviral vectors has gained increasing attention. This study investigates the effect of adenovirus-mediated gene transfer on endothelial cell (EC) count in human eye bank corneas. METHODS: A replication-defective adenoviral vector containing the gene for green fluorescent protein was used to transduce organ-cultured normal human eye bank and porcine corneas. Transgene expression and EC count were assessed by light and fluorescence microscopy. RESULTS: The transgene was expressed earlier by porcine EC (27% of all EC on day 2) than by human EC (6% on day 2), but the maximal number of EC finally expressing the transgene was higher in human than in porcine corneas (45 vs. 31% of all EC on day 12). Gene transfer caused considerably less EC loss in human than in porcine corneas (2 vs. 60% EC loss after 10 days). CONCLUSIONS: Adenoviral vectors for ex vivo gene transfer are more efficient and less toxic in normal human eye bank corneas than in porcine corneas, but human EC require more time until expression of transgenic proteins.


Asunto(s)
Adenoviridae/genética , Córnea/metabolismo , Expresión Génica/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Anciano , Anciano de 80 o más Años , Animales , Recuento de Células , Supervivencia Celular , Virus Defectuosos , Endotelio Corneal/patología , Bancos de Ojos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Fluorescente , Persona de Mediana Edad , Técnicas de Cultivo de Órganos , Porcinos , Donantes de Tejidos
20.
Hum Gene Ther ; 16(1): 35-48, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15703487

RESUMEN

Plasmid DNA encoding human interleukin 12 (IL-12) was produced under GMP conditions and injected into lesions of nine patients with malignant melanoma (stage IV) previously treated with both standard and nonstandard therapies. The treatment was based on efficacy in preclinical studies with melanoma in mice and gray horses. The DNA was applied in cycles, three injections per cycle, for up to seven cycles. Three therapy arms comprised low (2 mg), medium (4 mg), and high (10 to 20 mg) amounts of total DNA. The therapy was well tolerated. Three of nine patients experienced a clinical response: two stable disease and one complete remission. One patient receiving a low dose of DNA experienced a long-lasting stabilization of the disease for more than 3 years, whereas the other two responders received high doses of DNA. All patients but one (patient 9) experienced a transient response at the intratumoral injection site. Immunohistochemical staining of responder sections showed local reduction of angiogenesis and lymphocyte infiltrations. All patients, in particular the clinical and local responders (patients 3, 7, and 8), exhibited an antigen-specific immune response against MAGE-1 and MART-1, which in some cases preexisted. Biopsies of responders showed some increase in IL-12, IP-10, and IFN-(). Serum levels revealed fluctuations. The results show that intratumoral injection of DNA produced some beneficial clinical effect. DNA encoding a cytokine may be useful as a therapeutic or adjuvant against various human cancers.


Asunto(s)
ADN/administración & dosificación , Terapia Genética , Inmunoterapia , Interleucina-12/genética , Melanoma/terapia , Neoplasias Cutáneas/terapia , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias , Femenino , Humanos , Inyecciones Intralesiones , Interferón gamma/metabolismo , Antígeno MART-1 , Masculino , Melanoma/inmunología , Melanoma/secundario , Antígenos Específicos del Melanoma , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Plásmidos , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/secundario
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...