Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Virol Methods ; 310: 114614, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36084768

RESUMEN

The determination of the electrostatic charge of biological nanoparticles requires a purified, mono-disperse, and concentrated sample. Previous studies proofed an impact of the preparation protocol on the stability and electro-hydrodynamics of viruses, whereas commonly used methods are often complex and do not allow the required sample throughput. In the present study, the application of the (I) steric exclusion chromatography (SXC) for the Orf virus (ORFV) purification and subsequent physicochemical characterization was evaluated and compared to (II) SXC followed by centrifugal diafiltration and (III) sucrose cushion ultracentrifugation. The three methods were characterized in terms of protein removal, size distribution, infectious virus recovery, visual appearance, and electrophoretic mobility as a function of pH. All preparation techniques achieved a protein removal of more than 99 %, and (I) an infectious ORFV recovery of more than 85 %. Monodisperse samples were realized by (I) and (III). In summary, ORFV samples prepared by (I) and (III) displayed comparable quality. Additionally, (I) offered the shortest operation time and easy application. Based on the obtained data, the three procedures were ranked according to eight criteria of possible practical relevance, which delineate the potential of SXC as virus preparation method for physicochemical analysis.


Asunto(s)
Virus del Orf , Virus , Virión , Ultracentrifugación , Sacarosa
2.
Vaccines (Basel) ; 8(2)2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32531997

RESUMEN

The potency of viral vector-based vaccines depends on their ability to induce strong transgene-specific immune response without triggering anti-vector immunity. Previously, Orf virus (ORFV, Parapoxvirus) strain D1701-V was reported as a novel vector mediating protection against viral infections. The short-lived ORFV-specific immune response and the absence of virus neutralizing antibodies enables repeated immunizations and enhancement of humoral immune responses against the inserted antigens. However, only limited information exists about the D1701-V induced cellular immunity. In this study we employed major histocompatibility complex (MHC) ligandomics and immunogenicity analysis to identify ORFV-specific epitopes. Using liquid chromatography-tandem mass spectrometry we detected 36 ORFV-derived MHC I peptides, originating from various proteins. Stimulated splenocytes from ORFV-immunized mice did not exhibit specific CD8+ T cell responses against the tested peptides. In contrast, immunization with ovalbumin-expressing ORFV recombinant elicited strong SIINFEKL-specific CD8+ T lymphocyte response. In conclusion, our data indicate that cellular immunity to the ORFV vector is negligible, while strong CD8+ T cell response is induced against the inserted transgene. These results further emphasize the ORFV strain D1701-V as an attractive vector for vaccine development. Moreover, the presented experiments describe prerequisites for the selection of T cell epitopes exploitable for generation of ORFV-based vaccines by reverse genetics.

3.
Viruses ; 11(2)2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30704093

RESUMEN

The Orf virus (ORFV; Parapoxvirus) strain D1701 with an attenuated phenotype and excellent immunogenic capacity is successfully used for the generation of recombinant vaccines against different viral infections. Adaption for growth in Vero cells was accompanied by additional major genomic changes resulting in ORFV strain variant D1701-V. In this study, restriction enzyme mapping, blot hybridization and DNA sequencing of the deleted region s (A, AT and D) in comparison to the predecessor strain D1701-B revealed the loss of 7 open reading frames (ORF008, ORF101, ORF102, ORF114, ORF115, ORF116, ORF117). The suitability of deletion site D for expression of foreign genes is demonstrated using novel synthetic early promoter eP1 and eP2. Comparison of promoter strength showed that the original vegf-e promoter Pv as well as promoter eP2 display an up to 11-fold stronger expression than promoter eP1, irrespective of the insertion site. Successful integration and expression of the fluorescent marker genes is demonstrated by gene- and insertion-site specific PCR assays, fluorescence microscopy and flow cytometry. For the first time ORFV recombinants are generated simultaneously expressing transgenes in two different insertion loci. That allows production of polyvalent vaccines containing several antigens against one or different pathogens in a single vectored ORFV vaccine.


Asunto(s)
Adaptación Fisiológica/genética , Genoma Viral , Virus del Orf/genética , Recombinación Genética , Transgenes , Animales , Chlorocebus aethiops , Eliminación de Gen , Vectores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Virus del Orf/fisiología , Células Vero
4.
Methods Mol Biol ; 1349: 177-200, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26458837

RESUMEN

Orf virus (ORFV) is an epitheliotropic poxvirus, which belongs to the genus Parapoxvirus. Among them the highly attenuated, apathogenic strain D1701-V is regarded as a promising candidate for novel virus vector vaccines. Our recent work demonstrated that those ORFV-based recombinants were able to induce protective, long-lasting immunity in various hosts that are non-permissive for ORFV. In this chapter we describe procedures for the generation, selection, propagation, and titration of ORFV recombinants as well as transgene detection by PCR or immunohistochemical staining.


Asunto(s)
Anticuerpos Antivirales/inmunología , Virus del Orf/genética , Vacunas Virales/genética , Anticuerpos Antivirales/genética , Vectores Genéticos , Humanos , Virus del Orf/inmunología , Parapoxvirus/genética , Parapoxvirus/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico , Proteínas Virales/genética , Proteínas Virales/inmunología , Vacunas Virales/biosíntesis
5.
Virus Genes ; 49(1): 68-79, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24743940

RESUMEN

Orf virus (ORFV) strain D1701-V, a Parapoxvirus belonging to the family Poxviridae, became attractive as a novel virus vector system that we successfully used for the generation of recombinant vaccines. Therefore, the identification of viral genes involved in host tropisms or immune modulation is of great interest, as for instance the ORFV-encoded ankyrin-repeat (AR) containing proteins. The present study shows for the first time that the ANK-1 designated gene product of ORFV126 is targeted to mitochondria of ORFV-infected and in ANK-1 transiently expressing cells. Taking advantage of ANK-1 EGFP fusion proteins and confocal fluorescence microscopy mutational and deletion analyses indicated the importance of AR8 and AR9, which may contain a novel class of mitochondria-targeting sequence (MTS) in the central to C-terminal part of this AR-containing protein. The fluorescent findings were corroborated by cell fractionation and Western blotting experiments. The presented results open the avenue for more detailed investigations on cellular binding partners and the function of ANK-1 in viral replication or virulence.


Asunto(s)
Ancirinas/análisis , Mitocondrias/química , Virus del Orf/fisiología , Proteínas Virales/análisis , Animales , Repetición de Anquirina , Ancirinas/genética , Fusión Artificial Génica , Chlorocebus aethiops , Análisis Mutacional de ADN , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Microscopía Confocal , Microscopía Fluorescente , Transporte de Proteínas , Células Vero , Proteínas Virales/genética
6.
PLoS One ; 8(12): e83802, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376753

RESUMEN

Previously we demonstrated the versatile utility of the Parapoxvirus Orf virus (ORFV) as a vector platform for the development of potent recombinant vaccines. In this study we present the generation of new ORFV recombinants expressing the hemagglutinin (HA) or nucleoprotein (NP) of the highly pathogenic avian influenza virus (HPAIV) H5N1. Correct foreign gene expression was examined in vitro by immunofluorescence, Western blotting and flow cytometry. The protective potential of both recombinants was evaluated in the mouse challenge model. Despite adequate expression of NP, the recombinant D1701-V-NPh5 completely failed to protect mice from lethal challenge. However, the H5 HA-expressing recombinant D1701-V-HAh5n mediated solid protection in a dose-dependent manner. Two intramuscular (i.m.) injections of the HA-expressing recombinant protected all animals from lethal HPAIV infection without loss of body weight. Notably, the immunized mice resisted cross-clade H5N1 and heterologous H1N1 (strain PR8) influenza virus challenge. In vivo antibody-mediated depletion of CD4-positive and/or CD8-posititve T-cell subpopulations during immunization and/or challenge infection implicated the relevance of CD4-positive T-cells for induction of protective immunity by D1701-V-HAh5n, whereas the absence of CD8-positive T-cells did not significantly influence protection. In summary, this study validates the potential of the ORFV vectored vaccines also to combat HPAIV.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Virus del Orf/genética , Vacunas Virales/genética , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , ADN Recombinante/genética , Expresión Génica , Inmunización , Ratones , Nucleoproteínas/genética , Nucleoproteínas/inmunología , Subgrupos de Linfocitos T/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Células Vero
7.
J Virol ; 87(3): 1618-30, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23175365

RESUMEN

The present study describes the generation of a new Orf virus (ORFV) recombinant, D1701-V-RabG, expressing the rabies virus (RABV) glycoprotein that is correctly presented on the surface of infected cells without the need of replication or production of infectious recombinant virus. One single immunization with recombinant ORFV can stimulate high RABV-specific virus-neutralizing antibody (VNA) titers in mice, cats, and dogs, representing all nonpermissive hosts for the ORFV vector. The protective immune response against severe lethal challenge infection was analyzed in detail in mice using different dosages, numbers, and routes for immunization with the ORFV recombinant. Long-term levels of VNA could be elicited that remained greater than 0.5 IU per ml serum, indicative for the protective status. Single applications of higher doses (10(7) PFU) can be sufficient to confer complete protection against intracranial (i.c.) challenge, whereas booster immunization was needed for protection by the application of lower dosages. Anamnestic immune responses were achieved by each of the seven tested routes of inoculation, including oral application. Finally, in vivo antibody-mediated depletion of CD4-positive and/or CD8-posititve T cell subpopulations during immunization and/or challenge infection attested the importance of CD4 T cells for the induction of protective immunity by D1701-V-RabG. This report demonstrates another example of the potential of the ORFV vector and also indicates the capability of the new recombinant for vaccination of animals.


Asunto(s)
Antígenos Virales/inmunología , Portadores de Fármacos , Vectores Genéticos , Glicoproteínas/inmunología , Virus del Orf/genética , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Linfocitos T CD4-Positivos/inmunología , Gatos , Modelos Animales de Enfermedad , Perros , Femenino , Glicoproteínas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Rabia/prevención & control , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/genética , Virus de la Rabia/genética , Análisis de Supervivencia , Proteínas del Envoltorio Viral/genética
8.
BMC Vet Res ; 8: 114, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22809544

RESUMEN

BACKGROUND: The Orf virus (ORFV), a zoonotic Parapoxvirus, causes pustular skin lesions in small ruminants (goat and sheep). Intriguingly, ORFV can repeatedly infect its host, despite the induction of a specific immunity. These immune modulating and immune evading properties are still unexplained. RESULTS: Here, we describe that ORFV infection of permissive cells impairs the intracellular transport of MHC class I molecules (MHC I) as a result of structural disruption and fragmentation of the Golgi apparatus. Depending on the duration of infection, we observed a pronounced co-localization of MHC I and COP-I vesicular structures as well as a reduction of MHC I surface expression of up to 50%. These subversion processes are associated with early ORFV gene expression and are accompanied by disturbed carbohydrate trimming of post-ER MHC I. The MHC I population remaining on the cell surface shows an extended half-life, an effect that might be partially controlled also by late ORFV genes. CONCLUSIONS: The presented data demonstrate that ORFV down-regulates MHC I surface expression in infected cells by targeting the late vesicular export machinery and the structure and function of the Golgi apparatus, which might aid to escape cellular immune recognition.


Asunto(s)
Ectima Contagioso/virología , Regulación de la Expresión Génica/fisiología , Genes MHC Clase I/fisiología , Aparato de Golgi/fisiología , Virus del Orf/fisiología , Animales , Anticuerpos Monoclonales , Chlorocebus aethiops , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Genes MHC Clase I/genética , Transporte de Proteínas , Células Vero
9.
Vaccine ; 29(49): 9256-64, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22001119

RESUMEN

This report describes the generation of a new recombinant Orf virus (ORFV; Parapoxvirus) expressing the major capsid protein VP1 (VP60) of the calicivirus, rabbit hemorrhagic disease virus (RHDV). Authentic expression of VP1 could be demonstrated in cells infected with the recombinant D1701-V-VP1 without the need for production of infectious ORFV progeny. Notably, infected cells also released empty calicivirus-like particles (VLPs). Challenge experiments showed that even a single immunization with ≥10(5) PFU of D1701-V-VP1 protected rabbits against lethal RHDV infection. ELISA tests indicated that the protective immunity mediated by D1701-V-VP1 did not strictly depend on the presence of detectable RHDV-specific serum antibodies. The induction of interleukin-2 found only in the sera of rabbits immunized with the D1701-V-VP1, but not in sera of rabbits immunized with the inactivated commercial vaccine RIKA-VACC, might indicate also some involvement of T-cells in protection. Collectively, this work adds another example of the successful use of the ORFV vector system for the generation of a recombinant vaccine, and demonstrates its potential as an alternative vaccine to protect rabbits against RHDV infection.


Asunto(s)
Infecciones por Caliciviridae/veterinaria , Virus de la Enfermedad Hemorrágica del Conejo/inmunología , Virus del Orf/inmunología , Proteínas Estructurales Virales/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Formación de Anticuerpos , Infecciones por Caliciviridae/prevención & control , Chlorocebus aethiops , ADN Viral/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos , Masculino , Virus del Orf/genética , Conejos , Células Vero , Ensayo de Placa Viral , Proteínas Estructurales Virales/genética
10.
Vaccine ; 27(27): 3584-91, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19464538

RESUMEN

The present study demonstrates the protective potential of novel baculovirus recombinants, which express the glycoproteins gB, gC, or gD of Pseudorabies virus (PRV; Alphaherpesvirus of swine) and additionally contain the glycoprotein G of Vesicular Stomatitis Virus (VSV-G) in the virion (Bac-G-PRV). To evaluate the protective capacity, mixtures of equal amounts of the PRV gB-, gC-, and gD-expressing baculoviruses were used for immunization. Three intramuscular immunizations with that Bac-G-PRV mixture could protect mice against a lethal PRV challenge infection. To achieve complete protection high titers of Bac-G-PRV and three immunizations were necessary. This immunization with Bac-G-PRV resulted in the induction of high titers of PRV-specific serum antibodies of the IgG2a subclass and of interferon (IFN)-gamma, indicating a Th1-type immune response. Moreover, splenocytes of immunized mice exhibited natural killer cell activity accompanied by the production of IFN-alpha and IFN-gamma. Collectively, the presented data demonstrate for the first time that co-expression of VSV-G in baculovirus recombinant vaccines can improve the induction of a protective immune response against foreign antigens.


Asunto(s)
Baculoviridae/genética , Herpesvirus Suido 1/inmunología , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Interferón gamma/biosíntesis , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas del Envoltorio Viral/genética
11.
Vaccine ; 24(37-39): 6256-63, 2006 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-16814432

RESUMEN

The present study demonstrates the protective potential of a novel prime-boost vaccination strategy of pigs against lethal Pseudorabies virus (PRV; Herpes suid 1) infection. Animals were primed with Sindbis virus-derived plasmids that express viral glycoproteins gC and gD (gC- and gD-pSIN) and subsequently booster immunized with Orf virus (ORFV; Parapoxvirus) recombinants expressing gC and gD (D1701-VrVgC and -VrVgD). The prime-boost vaccination induced strong humoral and cellular-like PRV-specific immune responses. All prime-boost vaccinated pigs survived the lethal challenge infection without PRV-specific clinical symptoms and presented excellent body weight loss attenuation. Most notably, nasal shedding of challenge virus was reduced by more than about 3log(10), clearly reducing the risk of infection of non-immunized pigs.


Asunto(s)
Herpesvirus Suido 1/inmunología , Inmunización Secundaria/veterinaria , Virus del Orf/inmunología , Seudorrabia/prevención & control , Enfermedades de los Porcinos/prevención & control , Vacunas de ADN/uso terapéutico , Proteínas del Envoltorio Viral/inmunología , Animales , Herpesvirus Suido 1/genética , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-4/biosíntesis , Interleucina-4/genética , Interleucina-4/inmunología , Virus del Orf/genética , Seudorrabia/inmunología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Virus Sindbis/genética , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Células TH1/inmunología , Células Th2/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/genética
12.
J Virol ; 79(21): 13509-18, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16227271

RESUMEN

Borna disease virus (BDV) frequently causes meningoencephalitis and fatal neurological disease in young but not old mice of strain MRL. Disease does not result from the virus-induced destruction of infected neurons. Rather, it is mediated by H-2(k)-restricted antiviral CD8 T cells that recognize a peptide derived from the BDV nucleoprotein N. Persistent BDV infection in mice is not spontaneously cleared. We report here that N-specific vaccination can protect wild-type MRL mice but not mutant MRL mice lacking gamma interferon (IFN-gamma) from persistent infection with BDV. Furthermore, we observed a significant degree of resistance of old MRL mice to persistent BDV infection that depended on the presence of CD8 T cells. We found that virus initially infected hippocampal neurons around 2 weeks after intracerebral infection but was eventually cleared in most wild-type MRL mice. Unexpectedly, young as well as old IFN-gamma-deficient MRL mice were completely susceptible to infection with BDV. Moreover, neurons in the CA1 region of the hippocampus were severely damaged in most diseased IFN-gamma-deficient mice but not in wild-type mice. Furthermore, large numbers of eosinophils were present in the inflamed brains of IFN-gamma-deficient mice but not in those of wild-type mice, presumably because of increased intracerebral synthesis of interleukin-13 and the chemokines CCL1 and CCL11, which can attract eosinophils. These results demonstrate that IFN-gamma plays a central role in host resistance against infection of the central nervous system with BDV and in clearance of BDV from neurons. They further indicate that IFN-gamma may function as a neuroprotective factor that can limit the loss of neurons in the course of antiviral immune responses in the brain.


Asunto(s)
Enfermedad de Borna/inmunología , Enfermedad de Borna/prevención & control , Virus de la Enfermedad de Borna/inmunología , Virus de la Enfermedad de Borna/aislamiento & purificación , Linfocitos T CD8-positivos/inmunología , Interferón gamma/inmunología , Enfermedades del Sistema Nervioso/inmunología , Enfermedades del Sistema Nervioso/prevención & control , Vacunación , Vacunas Virales/administración & dosificación , Factores de Edad , Animales , Enfermedad de Borna/virología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Quimiocinas/inmunología , Pruebas Inmunológicas de Citotoxicidad , Eosinófilos/patología , Vectores Genéticos , Inyecciones Intramusculares , Interferón gamma/deficiencia , Interleucina-13/inmunología , Ratones , Ratones Noqueados , Enfermedades del Sistema Nervioso/virología , Proteínas de la Nucleocápside/biosíntesis , Proteínas de la Nucleocápside/genética , Parapoxvirus/genética , Bazo/inmunología , Vacunas Sintéticas/administración & dosificación , Virus Vaccinia/genética
13.
J Gen Virol ; 86(Pt 2): 399-403, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15659759

RESUMEN

Borna disease virus (BDV) can persistently infect the central nervous system and induce CD8+ T-cell-mediated neurological disease in MRL mice. To determine whether specific immune priming would prevent disease, a prime-boost immunization protocol was established in which intramuscular injection of a recombinant parapoxvirus expressing BDV nucleoprotein (BDV-N) was followed by intraperitoneal infection with vaccinia virus expressing BDV-N. Immunized wild-type and perforin-deficient mice remained healthy after intracerebral infection with BDV and contained almost no virus in the brain at 5 weeks post-challenge. Immunization failed to induce resistance against BDV in mice lacking mature CD8+ T cells. Immunization of perforin-deficient mice with a poxvirus vector expressing mutant BDV-N lacking the known CD8+ T-cell epitope did not efficiently block multiplication of BDV in the brain and did not prevent neurological disease, indicating that vaccine-induced immunity to BDV in wild-type and perforin-deficient mice resulted from the action of CD8+ T cells.


Asunto(s)
Enfermedad de Borna/prevención & control , Virus de la Enfermedad de Borna , Vacunación , Vacunas Virales/administración & dosificación , Animales , Enfermedad de Borna/inmunología , Enfermedad de Borna/virología , Virus de la Enfermedad de Borna/genética , Virus de la Enfermedad de Borna/inmunología , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Vectores Genéticos , Inyecciones Intramusculares , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Mutación , Nucleoproteínas/biosíntesis , Nucleoproteínas/genética , Parapoxvirus/genética , Perforina , Proteínas Citotóxicas Formadoras de Poros , Vacunas Sintéticas/administración & dosificación , Proteínas Virales/biosíntesis , Proteínas Virales/genética
14.
J Virol ; 79(1): 314-25, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15596826

RESUMEN

The Parapoxvirus Orf virus represents a promising candidate for novel vector vaccines due to its immune modulating properties even in nonpermissive hosts such as mouse or rat. The highly attenuated Orf virus strain D1701 was used to generate a recombinant virus (D1701-VrVp40) expressing nucleoprotein p40 of Borna disease virus, which represents a major antigen for the induction of a Borna disease virus-specific humoral and cellular immune response. Infection with Borna disease virus leads to distinct neurological symptoms mediated by the invasion of activated specific CD8+ T cells into the infected brain. Usually, Borna disease virus is not cleared from the brain but rather persists in neural cells. In the present study we show for the first time that intramuscular application of the D1701-VrVp40 recombinant protected rats against Borna disease, and importantly, virus clearance from the infected brain was demonstrated in immunized animals. Even 4 and 8 months after the last immunization, all immunized animals were still protected against the disease. Initial characterization of the immune cells attracted to the infected brain areas suggested that D1701-VrVp40 mediated induction of B cells and antibody-producing plasma cells as well as T cells. These findings suggest the induction of various defense mechanisms against Borna disease virus. First studies on the role of antiviral cytokines indicated that D1701-VrVp40 immunization did not lead to an enhanced early response of gamma or alpha interferon or tumor necrosis factor alpha. Collectively, this study describes the potential of the Orf virus vector system in mediating long-lasting, protective antiviral immunity and eliminating this persistent virus infection without provoking massive neuronal damage.


Asunto(s)
Enfermedad de Borna/prevención & control , Virus de la Enfermedad de Borna/inmunología , Virus del Orf/genética , Proteínas Virales/inmunología , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Antivirales/sangre , Enfermedad de Borna/inmunología , Enfermedad de Borna/fisiopatología , Enfermedad de Borna/virología , Virus de la Enfermedad de Borna/fisiología , Encéfalo/virología , Línea Celular , Citocinas/metabolismo , Cobayas , Inmunización , Virus del Orf/metabolismo , ARN Viral/análisis , Ratas , Ratas Endogámicas Lew , Recombinación Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Vacunas Virales/genética , Vacunas Virales/inmunología
15.
Vet Immunol Immunopathol ; 102(4): 389-97, 2004 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-15541792

RESUMEN

In Chinese Meishan/German Landrace cross-bred swine F2 generation interferon gamma (IFN-gamma) production by peripheral blood mononuclear cells (PBMC) was determined directly ex vivo at different time points after survival of a virulent pseudorabies virus (PRV) infection. This reactivity was compared with the reactivity of naïve PBMC. Significant IFN-gamma production was determined in ELISA and ELISPOT only after in vitro PBMC re-stimulation with PRV and not with the closely related bovine herpesvirus BHV-1. The PRV-specific IFN-gamma secretion from re-stimulated PBMC showed high levels 6 days after infection, before the presence of serum antibodies, and it persisted at a high level over a 3 months period. The response of a group of eight piglets infected intranasally with PRV varied. Only two animals showed the expected typical fever response. PRV specific IFN-gamma production by PBMC clearly indicated that infection had occurred. Early significant IFN-gamma production by primed PBMC turned out to be a reliable and specific ex vivo marker for cellular response against productive PRV infection in swine before antibody formation.


Asunto(s)
Interferón gamma/biosíntesis , Leucocitos Mononucleares/inmunología , Seudorrabia/inmunología , Enfermedades de los Porcinos/inmunología , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Ensayo de Inmunoadsorción Enzimática/veterinaria , Herpesvirus Suido 1/inmunología , Porcinos/inmunología , Enfermedades de los Porcinos/virología
16.
Oncogene ; 23(39): 6639-46, 2004 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-15235583

RESUMEN

Alterations in signalling via the Raf/MEK/ERK pathway interfere with influenza A virus replication in cell culture. While virus yields are reduced in cells expressing dominant-negative Raf or ERK, virus propagation is enhanced upon expression of constitutively active Raf or MEK. To study the impact of active Raf on influenza virus propagation in vivo, we investigated transgenic mice expressing an activated mutant of c-Raf (Raf-BxB) in the main target tissue of influenza virus, the lung. Raf-BxB expression results in multicentric alveolar adenomas. Influenza virus A infection of Raf-BxB mice results in increased disease symptoms and higher mortality rates. The immune response against viral pathogens in transgenic animals did not differ from wild-type mice as determined by the use of a Pseudorabies virus (PRV) as a model for a viral infection not affecting the lung. No significant differences of influenza virus titers in the lung of Raf-BxB and wild-type mice were observed. However, immunohistology revealed increased numbers of influenza NP-positive cells in the alveolar linings of Raf-BxB mice, demonstrating the strong tropism of influenza virus for cells expressing active Raf. These findings disclose the possibility to use modified influenza virus for the therapy of tumors with an activated Ras/Raf signalling pathway.


Asunto(s)
Virus de la Influenza A/patogenicidad , Pulmón/enzimología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Línea Celular , Perros , Virus de la Influenza A/fisiología , Ratones , Especificidad de la Especie , Tasa de Supervivencia , Replicación Viral
17.
J Virol ; 77(17): 9312-23, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12915547

RESUMEN

Orf virus (ORFV; Parapoxvirus ovis) was used to develop a novel vector system for the generation of effective and safe live vaccines. Based on the attenuated ORFV strain D1701-V, recombinants were produced that express the glycoproteins gC (D1701-VrVgC) or gD (D1701-VrVgD) of the alphaherpesvirus of swine, pseudorabies virus (PRV). Expression of gC and gD was also demonstrated on the surface of recombinant virus-infected murine cells that do not produce infectious ORFV. Single or combined immunization with the ORFV recombinants protected different mouse strains of a host species nonpermissive for ORFV against a fulminant, lethal PRV challenge infection equal to immunization with PRV live vaccine. Most notably, even a single immunization with D1701-VrVgC was protective, whereas two applications of D1701-VrVgD were required for immune protection. The higher protective capacity of D1701-VrVgC correlated with the induction of a strong specific humoral immune response. This suggestion was supported by transfer experiments using sera from recombinant-immunized mice, which resulted in partial gC but not gD antibody-mediated protection of the naïve recipients. Remarkably, immunization of different immune-deficient mice demonstrated that the application of the PRV gC-expressing recombinant controlled the challenge infection in the absence of either CD4(+) or CD8(+) T cells, B cells, or an intact perforin pathway. In contrast, D1701-VrVgD-immunized mice lacking CD4(+) T cells exhibited reduced protection, whereas animals lacking CD8(+) T cells, B cells, or perforin resisted the challenge infection. The present study demonstrates the potential of these new vector vaccines to efficiently prime both protective humoral and cell-mediated immune mechanisms in a host species nonpermissive for the vector virus.


Asunto(s)
Vectores Genéticos , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/inmunología , Parapoxvirus/genética , Animales , Anticuerpos Antivirales/biosíntesis , Genoma Viral , Inmunidad Celular , Inmunización Pasiva , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Seudorrabia/inmunología , Seudorrabia/prevención & control , Recombinación Genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/farmacología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/genética , Vacunas Virales/farmacología
18.
Virus Genes ; 26(1): 25-30, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12680690

RESUMEN

The infection of humans with Boma disease virus (BDV) is still a matter of debate. In a recent publication, we described a BDV (RW98) isolated from the blood of a psychiatric patient. The RNA of this virus differed more than 5% from that of the widely used strain He/80, which was supposed to represent our laboratory virus. Here, we show that the virus used in our laboratory was not He/80 and, furthermore, that RW98 has sequence identity to the laboratory strain. We also present data that BDV-specific nucleic acid detected in blood of the donor of the presumed RW98 isolate and one other patient differs from all known BDV-p24 sequences, arguing for the existence of BDV sequences in man.


Asunto(s)
Enfermedad de Borna/virología , Virus de la Enfermedad de Borna/genética , ARN Viral/genética , Viremia/virología , Animales , Secuencia de Bases , Virus de la Enfermedad de Borna/aislamiento & purificación , Línea Celular , Patos/virología , Síndrome de Fatiga Crónica/virología , Humanos , Mamíferos/virología , Trastornos Mentales/virología , Datos de Secuencia Molecular , Nucleoproteínas/genética , Fosfoproteínas/genética , Filogenia , ARN Viral/sangre , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Proteínas Virales/genética
19.
Virology ; 308(2): 317-29, 2003 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-12706081

RESUMEN

A novel porcine gammaherpesvirus was detected in the blood of domestic pigs by PCR. With degenerate-primer PCR and subsequent long-distance PCR approaches a 60-kbp genome stretch was amplified. Sequence analysis revealed the presence of the gammaherpesvirus ORFs 03 to 46 as well as a putative chemokine receptor and a v-bcl-2 gene. The 60-kbp sequence was compared with the corresponding sequence of the porcine lymphotropic herpesvirus 1 (PLHV-1) published recently and the sequence of PLHV-2, which was amplified from porcine tonsil. Considerable sequence differences (amino acid identities: 49-89%) were found between the novel virus and PLHV-1 as well as PLHV-2, which were very closely related to each other (amino acid identities: 85-98%). The novel virus had essentially the same genome organization as PLHV-1 and -2 and was therefore designated PLHV-3. Like PLHV-1 and -2, PLHV-3 was frequently found in the blood and in lymphoid organs of domestic and feral pigs from different geographic locations. In the blood, the PLHVs were detected predominantly in B-cells. Indication for latent as well as productive PLHV-3 infection was found in the porcine B-cell line L23. It can be concluded that the PLHVs are widespread and are likely to cause a persistent B-lymphotropic infection. Since PLHV-1 has been implicated in the development of porcine posttransplantation lymphoproliferative disease, all porcine lymphotropic gammaherpesviruses are of concern when pigs are used as donors in xenotransplantation.


Asunto(s)
Gammaherpesvirinae/genética , Animales , Linfocitos B/virología , Línea Celular , Genoma Viral , Leucocitos Mononucleares/virología , Sistemas de Lectura Abierta , Reacción en Cadena de la Polimerasa , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...