Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Schweiz Arch Tierheilkd ; 161(1): 23-31, 2019 Jan.
Artículo en Alemán | MEDLINE | ID: mdl-30602429

RESUMEN

INTRODUCTION: European hedgehogs (Erinaceus europaeus) have a high exposure to various ticks, which could transmit pathogens with direct health significance for the host and may have zoonotic potential. Tick-borne meningoencephalitis (FSME) is an important tick-borne disease in Switzerland, caused by the tick-borne encephalitis virus. About its occurrence in the European hedgehog population is little known. The present study examined various organs, blood and ticks of 65 European hedgehogs to obtain data of FSME virus presence in this species in Switzerland. Real-time RT-PCR from the lungs, liver, spleen and kidney of 56 hedgehogs and of 114 infesting ticks (Ixodes hexagonus or Ixodes ricinus) were used for the detection of viral RNA. In addition, 19 blood samples were tested for antibodies against FSME by ELISA. FSME virus antibodies were detected for the first time in the serum of a European hedgehog. Lung and spleen tissue samples of the same animal tested also weak virus positive on RT-PCR. Clinically, the hedgehog showed neurological symptoms, although these symptoms could have originated from an other diseases. No viral RNA was detected in any of the ticks. This study could not confirm if the meningoencephalitis in the hedgehog was triggered by the FSME viral infection. Nevertheless, the simultaneous detection of antibodies and virus RNA in the same animal makes the European hedgehog a competent host of the tick-borne encephalitis virus and leads to the assumption that this species can act as a reservoir.


INTRODUCTION: En raison du nombre élevé de tiques présents chez les hérissons d'Europe (Erinaceus europaeus), ces animaux sont fortement exposés aux différents pathogènes qu'ils transmettent, pathogènes qui, en plus de l'importance directe pour la santé de l'hôte, peuvent aussi avoir un potentiel en termes de zoonose. La méningo-encéphalite à tique est, en Suisse, une maladie importante transmise par les tiques. Elle est causée par le virus de la méningo-encéphalite verno-estivale. Son occurrence chez les hérissons d'Europe est jusqu'à maintenant peu connue. Au travers de l'étude des organes, du sang et des tiques provenant de 65 hérissons européens, il devrait pour la première fois être possible de se prononcer sur la présence du virus chez cette espèce en Suisse. La détection de l'ARN viral a été effectuée au moyen d'une RT-PCR en temps réel sur les poumons, le foie, la rate et les reins de 56 hérissons ainsi que sur un total de 114 tiques dont ils étaient porteurs, appartenant aux espèces Ixodes hexagonus ou Ixodes ricinus. En outre, 19 échantillons de sang ont été testés par ELISA pour des anticorps contre le virus. Dans la présente étude, des anticorps contre le virus de l'encéphalite à tiques dans le sérum d'un hérisson européen ont pu être détectés pour la première fois. Les échantillons de poumon et de rate du même animal ont également montré une faible présence virale. Le même hérisson a présenté des symptômes neurologiques, mais ceux-ci pouvaient également être associés à d'autres maladies. On n'a démontré la présence d'ARN viral chez aucune tique. La possibilité d'une encéphalite causée par l'infection virale chez les hérissons ne peut pas être confirmée ou exclues avec cette étude. La détection simultanée des anticorps et de l'ARN viral chez le même animal fait du hérisson européen non seulement un hôte compétent du virus de l'encéphalite verno-estivale mais donne également également à penser que cette espèce pourrait servir de réservoir.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/aislamiento & purificación , Encefalitis Transmitida por Garrapatas/veterinaria , Erizos/virología , Meningoencefalitis/veterinaria , Animales , Anticuerpos Antivirales/sangre , Vectores Arácnidos/virología , Reservorios de Enfermedades/veterinaria , Reservorios de Enfermedades/virología , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/epidemiología , Encefalitis Transmitida por Garrapatas/virología , Erizos/parasitología , Inmunoglobulina G/sangre , Ixodes/virología , Masculino , Meningoencefalitis/epidemiología , Meningoencefalitis/virología , ARN Viral/análisis , Suiza/epidemiología
2.
Gene Ther ; 22(5): 382-90, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25739989

RESUMEN

We have recently shown that targeted expression of myelin oligodendrocyte glycoprotein (MOG) to dendritic cells with self-inactivating-lentivirus vectors induces antigen-specific tolerance in naive antigen-specific CD4+ T cells and protects mice from experimental autoimmune encephalomyelitis (EAE). In the present study, we demonstrate that this approach also induces tolerance of activated antigen-specific CD4+ T cells and completely protects mice from passive EAE induction. Tolerance induction did not correlate with the depletion of the preactivated antigen-specific CD4+ T cells. However, upon isolation and in vitro re-stimulation at day 6 after adoptive transfer the MOG-specific CD4+ T cells from the non-tolerized mice produced large amounts of inflammatory cytokines, whereas those from tolerized mice did not. This unresponsiveness correlated with the upregulation of regulatory molecules associated with anergy and regulatory T cells (Tregs). The in vivo depletion of Tregs resulted in EAE susceptibility of the tolerized animals, suggesting that these cells have indeed a role in tolerance induction/maintenance.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Activación de Linfocitos , Traslado Adoptivo , Animales , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Regulación hacia Arriba
3.
Br J Cancer ; 110(1): 94-106, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24196790

RESUMEN

BACKGROUND: Emerging studies have shown the potential benefit of arming oncolytic viruses with therapeutic genes. However, most of these therapeutic genes are placed under the regulation of ubiquitous viral promoters. Our goal is to generate a safer yet potent oncolytic herpes simplex virus type-1 (HSV-1) for cancer therapy. METHODS: Using bacterial artificial chromosome (BAC) recombineering, a cell cycle-regulatable luciferase transgene cassette was replaced with the infected cell protein 6 (ICP6) coding region (encoded for UL39 or large subunit of ribonucleotide reductase) of the HSV-1 genome. These recombinant viruses, YE-PC8, were further tested for its proliferation-dependent luciferase gene expression. RESULTS: The ability of YE-PC8 to confer proliferation-dependent transgene expression was demonstrated by injecting similar amount of viruses into the tumour-bearing region of the brain and the contralateral normal brain parenchyma of the same mouse. The results showed enhanced levels of luciferase activities in the tumour region but not in the normal brain parenchyma. Similar findings were observed in YE-PC8-infected short-term human brain patient-derived glioma cells compared with normal human astrocytes. intratumoural injection of YE-PC8 viruses resulted in 77% and 80% of tumour regression in human glioma and human hepatocellular carcinoma xenografts, respectively. CONCLUSION: YE-PC8 viruses confer tumour selectivity in proliferating cells and may be developed further as a feasible approach to treat human cancers.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Herpesvirus Humano 1/fisiología , Viroterapia Oncolítica/métodos , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/virología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virología , Ciclo Celular/genética , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Glioma/genética , Glioma/virología , Herpesvirus Humano 1/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virología , Luciferasas/genética , Ratones , Ratones Desnudos , Ratones SCID , Elementos Reguladores de la Transcripción , Transcripción Genética , Transgenes , Células Vero , Proteínas Virales/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Gene Ther ; 20(5): 556-66, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22951454

RESUMEN

The aim of this work was to induce permanent tolerance toward self-antigens involved in autoimmune diseases, such as multiple sclerosis (MS). We hypothesized that the stable auto-antigen presentation by dendritic cells (DCs) would tolerize auto-reactive T cells and, therefore, prevent disease development in a mouse model of experimental autoimmune encephalomyelitis (EAE), which closely resembles MS. Specifically, our strategy included the ex vivo modification of hematopoietic stem cells (HSCs) with self-inactivating (SIN) lentivirus vectors that transcriptionally target the expression of myelin antigens to DCs. As SIN lentivirus vectors support the genomic integration of transgene sequences in HSC, the transduced and transplanted HSC may provide a constant supply of antigen expressing steady-state DCs. Here, we demonstrate that targeting myelin oligodendrocyte glycoprotein (MOG) expression to DCs indeed resulted in complete and stable protection from EAE. No histological signs of EAE, such as demyelination, axonal damage, or infiltration of leukocytes in brain, spinal cord and optical nerve, were observed in tolerized mice. Tolerance induction was concomitant with the efficient deletion of MOG-specific T cells and the generation of Foxp3(+) regulatory T cells and, most importantly, directed toward a specific self-antigen while T-cell reactivity to unrelated foreign antigens was fully preserved.


Asunto(s)
Autoantígenos , Células Dendríticas/metabolismo , Tolerancia Inmunológica/genética , Esclerosis Múltiple/terapia , Animales , Autoantígenos/genética , Autoantígenos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Modelos Animales de Enfermedad , Vectores Genéticos , Células Madre Hematopoyéticas/inmunología , Humanos , Lentivirus , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología
5.
Gene Ther ; 14(15): 1143-51, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17495946

RESUMEN

Standard treatment for glioblastoma multiforme and other brain tumors consists of surgical resection followed by combined radio-/chemotherapy. However, radiation resistance of tumor cells limits the success of this treatment, and the tumors invariably recur. Therefore, the selective inhibition of molecular mediators of radiation resistance may provide therapeutic benefit to the patient. One of these targets is the Rad51 protein, which is a key component of the homologous recombinational repair of DNA double-strand breaks. Here, we investigated whether post-transcriptional silencing of Rad51 by herpes simplex virus-type 1 (HSV-1) amplicon vector-mediated short interfering RNA expression can enhance the antitumor effect of radiation therapy. We demonstrate that these vectors specifically and efficiently inhibited the radiation-induced recruitment of Rad51 into nuclear foci in human glioma cells. The combination of vector-mediated silencing of Rad51 expression and treatment with ionizing radiation resulted in a pronounced reduction of the survival of human glioma cells in culture. In athymyc mice, a single intratumoral injection of Rad51-specific HSV-1 amplicon vector followed by a single radiation treatment resulted in a significant decrease in tumor size. In control animals, including mice that received an intratumoral injection of Rad51-specific amplicon vector but no radiation treatment, the tumor sizes increased.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glioma/terapia , Herpesvirus Humano 1/genética , Recombinasa Rad51/antagonistas & inhibidores , Animales , Apoptosis , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Terapia Combinada , Femenino , Expresión Génica , Silenciador del Gen , Vectores Genéticos/genética , Glioblastoma/terapia , Glioma/patología , Glioma/radioterapia , Humanos , Inyecciones Intralesiones , Ratones , Ratones Desnudos , Interferencia de ARN , ARN Interferente Pequeño/genética , Recombinasa Rad51/genética , Tolerancia a Radiación/fisiología , Resultado del Tratamiento
6.
Gene Ther ; 12(23): 1707-17, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16034459

RESUMEN

For leukemia vaccine generation, high-efficiency gene transfer is required to express immunomodulatory molecules that stimulate potent antileukemic immune responses. In this context, herpes simplex virus type-1 (HSV-1)-derived vectors have proven to be a promising tool for genetic modification of lymphoblastic leukemia cells. Yet, vector-associated viral protein expression might inadvertently modulate vaccine efficacy facilitating both immune evasion and immune stimulation. To explore the issue of immune-stimulation versus immune-suppression in immature lymphoblastic leukemia cells, two types of HSV-1 amplicon vectors, helper virus-dependent and helper virus-free that express the immunomodulatory molecules CD70 and IL-2, were compared with regard to their vector-associated immunomodulatory potential. We first established that lymphoblastic cell lines and primary acute lymphoblastic leukemia (ALL) cells express HSV receptor genes. Lymphoblastic cell lines were transduced with high efficiency, and in primary ALL cells high gene transfer rates of 47+/-15 and 42+/-14% were obtained with helper virus-dependent and -free HSV-1 amplicon vectors, respectively. The efficacy of the two amplicon vectors to induce antineoplastic responses was assessed in a vaccine setting in mice with pre-existing highly malignant lymphoblastic disease. Treatment of mice with vaccine cells transgenically expressing CD70+IL2 significantly suppressed lymphoblastic cell proliferation and improved survival. Of note, when helper virus-dependent HSV-1 amplicon vectors were used for vaccine preparation, the high immunogenic potential of the vector itself, in the absence of transgenic CD70+IL2 expression, seemed to be sufficient to mediate protection comparable to the antineoplastic response achieved by expression of immunomodulatory molecules. Thus for vaccine generation in B lymphoblastic leukemia, the immunogenic potential of HSV-1 helper virus-dependent amplicon vectors does provide additional benefit to the high transduction efficiency of HSV-1-derived vectors.


Asunto(s)
Vacunas contra el Cáncer/genética , Terapia Genética/métodos , Herpesvirus Humano 1/genética , Inmunoterapia/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Ligando CD27 , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Femenino , Expresión Génica , Genes MHC Clase I , Ingeniería Genética , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Transducción Genética/métodos , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología
7.
Gene Ther ; 12(16): 1283-8, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15889134

RESUMEN

The purpose of our study was to evaluate the transduction profiles of herpes simplex virus type 1 (HSV-1)-based amplicon vectors following subretinal injection in the rat. Two amplicon vectors were tested, pHy-CMVGFP and pHy-RPEGFP, both carrying the green fluorescent protein (GFP) under the control of the cytomegalovirus (CMV) ubiquitous promoter or the RPE65-specific promoter, respectively. For the two amplicon vectors, the GFP reporter gene was efficiently expressed in retinal pigment epithelial (RPE) cells but not in the adjacent photoreceptors. GFP expression was maximum as early as 2 days post-administration but decreased over time to become almost undetectable at 6 weeks postinjection. Super-transduction with a second amplicon vector, pHSVlac, reactivated expression of GFP in approximately 10% of the cells initially transduced at 2 days postinjection of pHy-CMVGFP or pHy-RPEGFP. Reactivation of transgene expression was transient, no GFP signal was detected 8 days after pHSVlac injection. In conclusion, HSV-1 amplicon vectors allow rapid and efficient, but transient, gene transfer in RPE cells following subretinal injection.


Asunto(s)
Proteínas del Ojo/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Herpesvirus Humano 1/genética , Retina/metabolismo , Transducción Genética/métodos , Animales , Proteínas Portadoras , Citomegalovirus/genética , Proteínas del Ojo/análisis , Proteínas del Ojo/metabolismo , Expresión Génica , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Inyecciones , Microscopía Fluorescente , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Enfermedades de la Retina/terapia , Factores de Tiempo , cis-trans-Isomerasas
8.
Gene Ther ; 12(7): 588-96, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15674397

RESUMEN

We investigated the variability in infectivity of cells in primary brain tumor samples from different patients using an HSV-1 amplicon vector. We studied the infectivity of HSV-1 amplicon vectors in tumor samples derived from neurosurgical resections of 20 patients. Cells were infected with a definite amount of HSV-1 amplicon vector HSV-GFP. Transduction efficiency in primary tumor cell cultures was compared to an established human glioma line. Moreover, duration of transgene expression was monitored in different tumor cell types. All primary cell cultures were infectable with HSV-GFP with variable transduction efficiencies ranging between 3.0 and 42.4% from reference human Gli36 Delta EGFR glioma cells. Transduction efficiency was significantly greater in anaplastic gliomas and meningiomas (26.7+/-17.4%) compared to more malignant tumor types (glioblastomas, metastases; 11.2+/-8.5%; P=0.05). To further investigate the possible underlying mechanism of this variability, nectin-1/HevC expression was analyzed and was found to contribute, at least in part, to this variability in infectability. The tumor cells expressed the exogenous gene for 7 to 61 days with significant shorter expression in glioblastomas (18+/-13 d) compared to anaplastic gliomas (42+/-24 d; P<0.05). Interindividual variability of infectivity by HSV-1 virions might explain, at least in part, why some patients enrolled in gene therapy for glioblastoma in the past exhibited a sustained response to HSV-1-based gene- and virus therapy. Infectivity of primary tumor samples from respective patients should be tested to enable the development of efficient and safe herpes vector-based gene and virus therapy for clinical application.


Asunto(s)
Neoplasias Encefálicas/virología , Vectores Genéticos , Herpesvirus Humano 1/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Moléculas de Adhesión Celular/metabolismo , Proliferación Celular , Expresión Génica , Herpesvirus Humano 1/patogenicidad , Humanos , Nectinas , Proteínas de Neoplasias/metabolismo , Receptores Virales/metabolismo , Factores de Tiempo , Transducción Genética , Células Tumorales Cultivadas
9.
Vet Microbiol ; 86(1-2): 103-13, 2002 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-11888694

RESUMEN

The most common viral disease of cats worldwide is the infection with feline herpesvirus 1 (FeHV-1). This infection may be followed by Herpetic stromal keratitis (HSK), which is supposed to have an immunopathological basis. Experiments using herpes simplex viruses (HSV) in mouse models indicated that HSK may be treated by topical application of the interleukin 10 (IL-10) gene. The objective of this study was the construction of human herpes simplex virus type 1 (HSV-1)-based amplicon vectors expressing feline interleukin genes and delivery of these genes into cells of feline origin. HSV-1-based amplicon vectors encoding either the enhanced green fluorescent protein, the feline IL-6 or the feline IL-10 under control of the HSV-1 immediate-early 4/5 promotor were constructed, packaged into amplicon particles, transduced into feline cells, and tested for RNA synthesis and biological activity. Feline cells were successfully transduced by HSV-1-based amplicon particles and RNA specific for the transgene was detected already at 2h post transduction, with a maximum at 24h. The recombinant feline IL-10 was functionally active as demonstrated by the reduction of both IL-12 p40 and interferon-gamma-mRNA production in Pansorbin stimulated feline peripheral mononuclear cells. Similarly, the recombinant feline IL-6, which was secreted into the supernatant of transduced cells, was able to support the growth of the IL-6-dependent murine B cell hybridoma 7TD1. HSV-1-based amplicon particles are able to transduce cells of feline origin with genes encoding biologically functional feline IL-10 or IL-6. It will be of high interest to study the effects of these tools in vivo.


Asunto(s)
Enfermedades de los Gatos/virología , Terapia Genética/veterinaria , Herpes Simple/veterinaria , Herpesvirus Humano 1/genética , Interleucina-10/genética , Interleucina-6/genética , Animales , Enfermedades de los Gatos/terapia , Gatos , Chlorocebus aethiops , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Herpes Simple/terapia , Herpesvirus Humano 1/química , Interferón gamma/biosíntesis , Interferón gamma/genética , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Interleucina-12/genética , Interleucina-6/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero
10.
Vet Microbiol ; 86(1-2): 165-74, 2002 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-11888699

RESUMEN

Herpes simplex virus-1 (HSV-1) based amplicon vectors are promising gene delivery vehicles because they have a large transgene capacity and can efficiently transduce many different cell types, including non-dividing cells, of various animal species. The Circ protein of bovine herpesvirus-1 (BHV-1) is a myristylated virion component of unknown function. Preliminary experiments with a circ gene deletion mutant indicated that Circ may influence the host's immune response by downregulating MHC-II expression in bovine monocytes. To get more insight into the function of Circ, amplicon vectors were constructed with various open reading frames (ORFs) under the control of the HSV-1 IE4/5 promoter: (i) the Circ ORF alone, (ii) a fusion ORF encoding an N-terminal Circ fused to the enhanced green fluorescent protein (eGFP), (iii) the eGFP ORF alone, and (iv) the Circ ORF in the inverted orientation. Upon helpervirus-free packaging into HSV-1 amplicon particles and transduction of Vero cells, both Circ alone and the Circ-eGFP fusion protein produced a punctate pattern within the cytoplasm, suggesting membrane association of the myristylated protein. In contrast, eGFP alone was evenly distributed over the cytoplasm of transduced cells. Upon infection of bovine buffy-coat cells, it was observed that cells of the monocyte lineage but not lymphocytes were transduced. Transgene expression reached a peak around 20h after transduction and lasted for at least 90h. Transduced monocytes underwent specific morphological changes, which may be attributed to Circ synthesis.


Asunto(s)
Terapia Genética/veterinaria , Herpesvirus Bovino 1/genética , Proteínas Virales/genética , Animales , Bovinos , Chlorocebus aethiops , Efecto Citopatogénico Viral/genética , Citometría de Flujo/veterinaria , Terapia Genética/métodos , Vectores Genéticos , Proteínas Fluorescentes Verdes , Herpesvirus Bovino 1/química , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Proteínas Luminiscentes/química , Monocitos , Plásmidos/genética , Transducción Genética/métodos , Células Vero , Proteínas Virales/química
11.
J Clin Microbiol ; 40(2): 519-23, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11825966

RESUMEN

A fluorogenic PCR was established for the quantification of feline herpesvirus 1 (FeHV-1) DNA in ocular fluid samples of clinically diseased cats. The new assay was specific for FeHV-1 and sensitive. The 100% detection rate ranged from 0.6 to 6 50% tissue culture infective doses per sample. When spiked samples with known quantities of virus were used, infectious virus titers and quantification of viral DNA by PCR correlated to each other in a linear fashion (R(2) = 0.9858) over a range of 4 orders of magnitude. Within this range, it was possible to calculate the FeHV-1 DNA content from a given infectious dose, and vice versa. The new diagnostic procedure was applied to ocular fluid samples from cats experimentally infected with FeHV-1 and specific FeHV-1-free cats. A good correlation between virus titer and quantitative PCR was observed, although only early in infection. In a second stage, the titer of infectious virus collapsed, while the PCR signal remained high. A constantly decreasing PCR signal accompanied by negative virus isolation was characteristic for a final stage of the infection. Finally, clinical samples from 20 cats that were suspected to suffer from FeHV-1 infection were analyzed. By comparing virus titers and quantitative PCR signals, it was possible to determine the current stage of the ongoing infection. Based on these findings, comparison of the results of consecutive samples allows the tracking of the course of the infection. Therefore, the new method combines the advantages of the two previously established conventional methods, qualitative PCR and virus isolation and titration.


Asunto(s)
Humor Acuoso/virología , Enfermedades de los Gatos/virología , ADN Viral/análisis , Herpesviridae/aislamiento & purificación , Reacción en Cadena de la Polimerasa/métodos , Animales , Gatos , Conjuntivitis Viral/veterinaria , Conjuntivitis Viral/virología , Herpesviridae/genética , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Queratitis Herpética/veterinaria , Queratitis Herpética/virología , Polimerasa Taq/metabolismo
12.
Cytometry ; 44(2): 93-9, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11378858

RESUMEN

BACKGROUND: In this study, we compared herpes simplex virus type 1 (HSV-1) amplicon vector stocks prepared by transient cotransfection with two different BAC-cloned packaging-defective HSV-1 helper genomes, fHSVDeltapacDelta27 and fHSVDeltapac, with respect to transduction efficiency and cytotoxicity. Both fHSVDeltapacDelta27 and fHSVDeltapac are packaging defective because the pac signals have been deleted; fHSVDeltapacDelta27 contains an additional deletion in the HSV-1 ICP27 gene, which increases the safety of the system. METHODS: HSV-1 amplicon pHSVGFP under the control of the HSV-1 immediate-early (IE) 4/5 promotor was packaged into virus particles by transient cotransfection with either fHSVDeltapacDelta27 or fHSVDeltapac DNA. Cultures were infected with the two different vector stocks and examined under the fluorescence microscope and analyzed by flow cytometry over a 5-day period to assess transduction efficiency and cytotoxicity. RESULTS: Both vector stocks, pHSVGFP[fHSVDeltapacDelta27] and pHSVGFP[fHSVDeltapac], efficiently transduced the target cells. Interestingly, the highest mean fluorescence intensities were measured at 1 day after infection, whereas the number of GFP-fluorescent cells reached a peak at day 3 after infection. At day 3 after infection, a slight increase in the number of dead cells was observed in those cultures transduced with high doses of vector stock. Between days 3 and 4 after infection, the number of dead cells increased dramatically in all the cultures, transduced and nontransduced. Only the cultures infected with a high dose of pHSVGFP[fHSVDeltapac] displayed a significant further increase in the number of dead cells between days 4 and 5 postinfection. CONCLUSIONS: Flow cytometry allowed comparison of transduction efficiency and cytotoxicity mediated by the two different amplicon vector stocks. Cultures infected with pHSVGFP[fHSVDeltapacDelta27] were more viable than those infected with pHSVGFP[fHSVDeltapac](P < 0.05). The practical implications of this study are at the level of vector design. Flow cytometry has proven a fast and reliable approach to assess the quality of potential gene transfer vectors prior to their use in (pre) clinical trials.


Asunto(s)
Vectores Genéticos/fisiología , Herpesvirus Humano 1/fisiología , Transducción Genética/métodos , Animales , Chlorocebus aethiops , Citometría de Flujo/métodos , Vectores Genéticos/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidad , Humanos , Células Vero
13.
Mol Ther ; 3(4): 591-601, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11319922

RESUMEN

Herpes simplex virus type 1 (HSV-1) amplicons are prokaryotic plasmids containing one or more transcriptional units and two cis-acting HSV-1 sequences: a viral origin of DNA replication and a viral DNA cleavage/packaging signal. In the presence of HSV-1 "helper" functions, amplicons are replicated and packaged into HSV-1 virions. Despite recent improvements in packaging methods, stocks of amplicon vectors are still contaminated with replication-competent helper virus at a frequency of 10(-4)-10(-6). To overcome this problem, we report that: (i) genetic modifications of HSV-1 genomes can be routinely achieved in Escherichia coli, either by homologous or site-specific recombination, (ii) a novel HSV-1 bacterial artificial chromosome (fHSVDeltapacDelta27 0+), which has a deletion in the essential gene encoding ICP27 and an addition of ICP0 "stuffer" sequences to increase its size to 178 kb, supports the replication and packaging of cotransfected amplicon DNA without generating replication-competent helper virus (<1 helper virus per 10(8) TU amplicon vectors), and (iii) the resulting amplicon stocks have titers of up to 3-10 x 10(8) TU/ml after concentration. Elimination of replication-competent helper virus from HSV-1 amplicon vector stocks further improves safety in gene transfer applications.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Virus Helper/genética , Herpesvirus Humano 1/genética , Simplexvirus/genética , Animales , Southern Blotting , Chlorocebus aethiops , Cósmidos/metabolismo , ADN/metabolismo , Escherichia coli/metabolismo , Eliminación de Gen , Modelos Genéticos , Mutagénesis Sitio-Dirigida , Plásmidos/metabolismo , Recombinación Genética , Células Vero
15.
J Gene Med ; 2(5): 379-89, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11045432

RESUMEN

BACKGROUND: This study investigates elements of herpes simplex virus type 1 (HSV-1) which influence transgene expression in tetracycline-regulated expression systems. METHODS: Different HSV-1 mutants were used to infect Vero cells that had been transfected with plasmids containing the luciferase gene under the control of tet-off or tet-on tetracycline-regulation systems. RESULTS: The baseline level of luciferase expression was elevated after infection with HSV-1 mutants lacking one or more immediate early genes encoding transactivating factors: ICP27, ICP4 and ICP0. With the tet-off system, not only was baseline expression elevated, but there was a complete loss of induction upon removal of tet when this regulatory system was brought into the cell by infection with helper virus-free amplicon vectors. Elevation of luciferase expression was also observed upon infection with the same HSV-1 mutants following transfection with a plasmid containing only a CMV minimal promoter driving luciferase (pUHC13-3). Only one HSV mutant (14Hdelta3), which bears a disruption in the transactivation domain of VP16 and is deleted for both ICP4 genes, did not increase baseline luciferase expression after transfection of pUHC13-3. The disregulating effects were dependent on virus dose and were not influenced by treatment with interferon (IFN)-alpha, which suppresses viral gene expression. Additional assays involving cotransfection of pUHC13-3 with a plasmid encoding of the HSV-1 transactivating factor ICP4 revealed that ICP4 was the most potent inducer of gene expression from the tetO/CMV minimal promoter. CONCLUSION: These results indicate that proteins encoded in the HSV-1 genome, especially the transactivating immediate early gene products (ICP4, ICP27 and ICP0) and the VP16 tegument protein can activate the tetO/ minimal CMV promoter and thereby interfere with the integrity of tetracycline-regulated transgene expression.


Asunto(s)
Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Herpesvirus Humano 1/genética , Tetraciclina/farmacología , Animales , Chlorocebus aethiops , Genes Inmediatos-Precoces , Proteínas Inmediatas-Precoces/genética , Luciferasas/genética , Mutación , Plásmidos/genética , Regiones Promotoras Genéticas , Transcripción Genética , Transfección , Células Vero , Proteínas Virales/metabolismo
17.
Hum Gene Ther ; 11(10): 1429-38, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10910140

RESUMEN

Subcutaneous vaccination therapy with glioma cells, which are retrovirally transduced to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF), has previously proven effective in C57BL/6 mice harboring intracerebral GL261 gliomas. However, clinical ex vivo gene therapy for human gliomas would be difficult, as transgene delivery via retroviral vectors occurs only in dividing cells and ex vivo glioma cells have a low growth fraction. To circumvent this problem, a helper virus-free herpes simplex virus type 1 (HSV-1) amplicon vector was used. When primary cultures of human glioblastoma cells were infected with HSV-1 amplicon vectors at an MOI of 1, more than 90% of both dividing and nondividing cells were transduced. When cells were infected with an amplicon vector, HSVGM, bearing the GM-CSF cDNA in the presence of Polybrene, GM-CSF secretion into the medium during the first 24 hr after infection was 1026 ng/10(6) cells, whereas mock-infected cells did not secrete detectable GM-CSF. Subcutaneous vaccination of C57BL/6 mice with 5 x 10(5) irradiated HSVGM-transduced GL261 cells 7 days prior to intracerebral implantation of 10(6) wild-type GL261 cells yielded 60% long-term survivors (>80 days), similar to the 50% long-term survivors obtained by vaccination with retrovirally GM-CSF-transduced GL261 cells. In contrast, animals vaccinated with the same number of nontranduced GL261 cells or with GL261 cells infected with helper virus-free packaged HSV-1 amplicon vectors carrying no transgene showed only 10% long-term survivors. In conclusion, helper virus-free HSV-1 amplicon vectors appear to be effective for cytokine-enhanced vaccination therapy of glioma, with the advantages that both dividing and nondividing tumor cells can be infected, no viral proteins are expressed, and these vectors are safe and compatible with clinical use.


Asunto(s)
Vacunas contra el Cáncer , Glioma/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Virus Helper/genética , Herpesvirus Humano 1/genética , Neoplasias Experimentales/terapia , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Femenino , Bromuro de Hexadimetrina/farmacología , Humanos , Operón Lac , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo , Transducción Genética , Transgenes/genética , Células Tumorales Cultivadas , Células Vero
18.
Am J Physiol Cell Physiol ; 278(3): C619-26, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10712251

RESUMEN

Analysis of RyR1 structure function in muscle cells is made difficult by the low (<5%) transfection efficiencies of myoblasts or myotubes using calcium phosphate or cationic lipid techniques. We inserted the full-length 15.3-kb RyR1 cDNA into a herpes simplex virus type 1 (HSV-1) amplicon vector, pHSVPrPUC between the ori/IE 4/5 promoter sequence and the HSV-1 DNA cleavage/packaging signal (pac). pHSVGN and pHSVGRyR1, two amplicons that expressed green fluorescent protein, were used for fluorescence-activated cell sorter analysis of transduction efficiency. All amplicons were packaged into HSV-1 virus particles using a helper virus-free packaging system and yielded 10(6) transducing vector units/ml. HSVRyR1, HSVGRyR1, and HSVGN virions efficiently transduced mouse myoblasts and myotubes, expressing the desired product in 70-90% of the cells at multiplicity of infection 5. The transduced cells appeared healthy and RyR1 produced by this method was targeted properly and restored skeletal excitation-contraction coupling in dyspedic myotubes. The myotubes produced sufficient protein to allow single-channel analyses from as few as 10 100-mm dishes. In most cases this method could preclude the need for permanent transfectants for the study of RyR1 structure function.


Asunto(s)
Técnicas de Transferencia de Gen , Herpesvirus Humano 1/genética , Músculo Esquelético/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Transfección/métodos , Animales , Animales Recién Nacidos , Calcio/farmacología , Diferenciación Celular , Células Cultivadas , Vectores Genéticos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/citología , Regiones Promotoras Genéticas , Proteínas Recombinantes/biosíntesis , Rianodina/farmacología , Canal Liberador de Calcio Receptor de Rianodina/genética
19.
Neurobiol Dis ; 6(5): 364-75, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10527804

RESUMEN

Huntington's disease (HD) is caused by an expanded glutamine tract, which confers a novel aggregation-promoting property on the 350-kDa huntingtin protein. Using specific antibodies, we have probed the structure of the polyglutamine segment in mutant huntingtin complexes formed in cell culture from either truncated or full-length protein. Complexes formed by a mutant amino terminal fragment most frequently entail a change in conformation that eliminates reactivity with the polyglutamine-specific mAb 1F8, coincident with production of insoluble aggregate. By contrast, complexes formed by the full-length mutant protein remain soluble and are invariably 1F8-reactive, indicating a soluble polyglutamine conformation. Therefore, aggregates in HD may form by different biochemical mechanisms that invoke different possibilities for the pathogenic process. If pathogenesis is triggered by a truncated fragment, it probably involves the formation of an insoluble aggregate. However, the observation of soluble complexes in which an HD-specific pathogenic conformation of the glutamine tract remains accessible suggests that pathogenesis could also be triggered at the level of full-length huntingtin by abnormal aggregation with normal or abnormal protein partners.


Asunto(s)
Cuerpo Estriado/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Péptidos/química , Animales , Anticuerpos Monoclonales , Línea Celular , Células Cultivadas , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Proteínas Nucleares/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Secuencias Repetitivas de Aminoácido , Solubilidad , Transfección
20.
Proc Natl Acad Sci U S A ; 96(22): 12697-702, 1999 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-10535985

RESUMEN

This study aimed to exploit bacterial artificial chromosomes (BAC) as large antigen-capacity DNA vaccines (BAC-VAC) against complex pathogens, such as herpes simplex virus 1 (HSV-1). The 152-kbp HSV-1 genome recently has been cloned as an F-plasmid-based BAC in Escherichia coli (fHSV), which can efficiently produce infectious virus progeny upon transfection into mammalian cells. A safe modification of fHSV, fHSVDeltapac, does not give rise to progeny virus because the signals necessary to package DNA into virions have been excluded. However, in mammalian cells fHSVDeltapac DNA can still replicate, express the HSV-1 genes, cause cytotoxic effects, and produce virus-like particles. Because these functions mimic the lytic cycle of the HSV-1 infection, fHSVDeltapac was expected to stimulate the immune system as efficiently as a modified live virus vaccine. To test this hypothesis, mice were immunized with fHSVDeltapac DNA applied intradermally by gold-particle bombardment, and the immune responses were compared with those induced by infection with disabled infectious single cycle HSV-1. Immunization with either fHSVDeltapac or disabled infectious single cycle HSV-1 induced the priming of HSV-1-specific cytotoxic T cells and the production of virus-specific antibodies and conferred protection against intracerebral injection of wild-type HSV-1 at a dose of 200 LD(50). Protection probably was cell-mediated, as transfer of serum from immunized mice did not protect naive animals. We conclude that BAC-VACs per se, or in combination with genetic elements that support replicative amplification of the DNA in the cell nucleus, represent a useful new generation of DNA-based vaccination strategies for many viral and nonviral antigens.


Asunto(s)
Cromosomas Bacterianos , Herpes Labial/prevención & control , Vacunas de ADN/inmunología , Animales , Biolística , Femenino , Herpes Labial/inmunología , Inmunoglobulina G/inmunología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/genética , Ensamble de Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...