Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Schweiz Arch Tierheilkd ; 161(1): 23-31, 2019 Jan.
Artigo em Alemão | MEDLINE | ID: mdl-30602429

RESUMO

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.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos/isolamento & purificação , Encefalite Transmitida por Carrapatos/veterinária , Ouriços/virologia , Meningoencefalite/veterinária , Animais , Anticorpos Antivirais/sangue , Vetores Aracnídeos/virologia , Reservatórios de Doenças/veterinária , Reservatórios de Doenças/virologia , Vírus da Encefalite Transmitidos por Carrapatos/genética , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/epidemiologia , Encefalite Transmitida por Carrapatos/virologia , Ouriços/parasitologia , Imunoglobulina G/sangue , Ixodes/virologia , Masculino , Meningoencefalite/epidemiologia , Meningoencefalite/virologia , RNA Viral/análise , Suíça/epidemiologia
2.
Gene Ther ; 22(5): 382-90, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25739989

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Ativação Linfocitária , Transferência Adotiva , Animais , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/prevenção & controle , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/imunologia , Fragmentos de Peptídeos/imunologia , Regulação para Cima
3.
Br J Cancer ; 110(1): 94-106, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24196790

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Herpesvirus Humano 1/fisiologia , Terapia Viral Oncolítica/métodos , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/virologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Glioma/genética , Glioma/virologia , Herpesvirus Humano 1/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virologia , Luciferases/genética , Camundongos , Camundongos Nus , Camundongos SCID , Elementos Reguladores de Transcrição , Transcrição Gênica , Transgenes , Células Vero , Proteínas Virais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Gene Ther ; 20(5): 556-66, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22951454

RESUMO

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.


Assuntos
Autoantígenos , Células Dendríticas/metabolismo , Tolerância Imunológica/genética , Esclerose Múltipla/terapia , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Modelos Animais de Doenças , Vetores Genéticos , Células-Tronco Hematopoéticas/imunologia , Humanos , Lentivirus , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Linfócitos T Reguladores/imunologia
5.
Nat Biotechnol ; 14(8): 1012-6, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9631042

RESUMO

To monitor the presence of introduced genes and the distribution of the encoded proteins in host tissues after gene transfer, we combined fluorescence in situ hybridization (FISH) and immunohistochemistry in two separate gene therapy paradigms. In brain tissue sections from animals injected with pHSVlac vector, we localized nuclei containing vector DNA both in cells expressing and not expressing beta-galactosidase (beta-gal). This suggests that the efficiency of gene transfer is affected not only by gene delivery, but also by cellular controls on gene expression. In a second paradigm, following myoblast transplantation, we detected donor nuclei in the muscle of a patient with Duchenne's muscular dystrophy. The donor nuclei were either surrounded by host nuclei or apparently fused in the patient's muscle fiber producing dystrophin. The combined FISH and immunohistochemistry assay offers greater sensitivity and more information than currently used polymerase chain reaction and protein detection methods.


Assuntos
Encéfalo/metabolismo , Protocolos Clínicos , Terapia Genética , Hibridização in Situ Fluorescente/métodos , Músculo Esquelético/metabolismo , Distrofias Musculares/terapia , Adulto , Animais , Distrofina/genética , Feminino , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/isolamento & purificação , Humanos , Masculino , Distrofias Musculares/genética , Ratos , Ratos Sprague-Dawley
6.
Hum Gene Ther ; 10(15): 2481-94, 1999 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-10543613

RESUMO

To improve gene transfer to CNS neurons, critical elements of herpes simplex virus 1 (HSV-1) amplicons and recombinant adeno-associated virus (AAV) vectors were combined to construct a hybrid amplicon vector, and then packaged via a helper virus-free system. We tested the HSV/AAV hybrid amplicon vectors for transduction efficiency and stability of transgene expression (green fluorescent protein) in primary neuronal cultures from rat fetal ventral mesencephalon, in comparison with traditional HSV amplicon, AAV, or adenovirus (Ad) vectors at the same multiplicity of infection. The HSA/AAV hybrid vectors transduced the highest number of primary neurons in culture 2 days after infection. As compared with all other vectors tested, only hybrid vectors containing the AAV rep gene maintained the 2-day level of transgene expression over 12 days in culture. This rep-containing hybrid vector was then tested for efficiency and safety in the brain. One month after injection into adult rat striatum (1 x 10(6) transducing units injected), transgene expression was observed within the striatum (ranging from 564 to 8610 cells) and the substantia nigra (via retrograde transport, ranging from 130 to 809 neurons). The HSV/AAV hybrid amplicon vectors transduced predominantly neurons within the striatum, and showed transduction efficacy similar to and in many cases higher than that of HSV amplicon vectors. No immune response was observed in the HSA/AAV hybrid vector-injected brains, as determined by immune markers specific for helper T lymphocytes, cytotoxic T lymphocytes, and microglia. This HSV/AAV hybrid system shows high transduction efficiency and stability in culture. The effective and safe transgene delivery into the nigrostriatal system illustrates its potential for therapeutic application for neurologic disorders, such as Parkinson and Huntington disease.


Assuntos
Corpo Estriado/metabolismo , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Simplexvirus/genética , Substância Negra/metabolismo , Animais , Biomarcadores , Células Cultivadas , Cruzamentos Genéticos , Ratos , Transdução Genética
7.
Hum Gene Ther ; 8(3): 359-70, 1997 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-9048203

RESUMO

Novel hybrid vectors, which incorporate critical elements of both herpes simplex virus type 1 (HSV-1) amplicon vectors and adeno-associated virus (AAV) vectors, are able to sustain transgene expression in dividing glioma cells for over 2 weeks. These vectors combine the high infectibility and large transgene capacity of HSV-1 vectors with the potential for episomal amplification and chromosomal integration of AAV vectors. The hybrid vectors contain the HSV-1 origin of DNA replication, oriS, and the DNA cleavage/packaging signal, pac, which allow amplicon replication and packaging in HSV-1 virions. The lacZ reporter gene under control of the CMV IE1 promoter is flanked by AAV inverted terminal repeat (ITR) sequences, which facilitate replication and genomic integration of this cassette in the host cell nucleus. Constructs were generated with or without the AAV rep gene (rep+ and rep-) to assess its importance in extending transgene expression. Expression of Rep proteins was confirmed by Western blot analysis. An HSV-1 amplicon construct containing the reporter gene, but no AAV sequences, was used as a control. Constructs were packaged into HSV-1 virions with or without helper virus and these vector stocks were used to infect human U87 glioma cells in culture. The hybrid vectors supported transgene retention and expression for over 2 weeks, whereas the control amplicon vector lost the transgene after 10 days. Expression was somewhat longer for the rep+ as compared to the rep- hybrid vectors. Toxicity due to the HSV-1 helper virus was eliminated using helper virus-free amplicon vector stocks. Transgene constructs could also be packaged in AAV virions, using AAV and adenovirus or HSV-1 helper functions. These HSV/AAV hybrid vectors should allow long-term, nontoxic gene delivery of DNA constructs to both dividing and nondividing cells.


Assuntos
Proteínas de Ligação a DNA/genética , Genes Virais , Vetores Genéticos , Glioma/genética , Herpesvirus Humano 1/genética , Proteínas Virais/genética , Animais , Western Blotting , Chlorocebus aethiops , Proteínas de Ligação a DNA/biossíntese , Dependovirus/genética , Expressão Gênica , Técnicas de Transferência de Genes , Genes Reporter , Glioma/terapia , Glioma/virologia , Humanos , Reação em Cadeia da Polimerase , Células Vero , Proteínas Virais/biossíntese
8.
Hum Gene Ther ; 11(10): 1429-38, 2000 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910140

RESUMO

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.


Assuntos
Vacinas Anticâncer , Glioma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Vírus Auxiliares/genética , Herpesvirus Humano 1/genética , Neoplasias Experimentais/terapia , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Brometo de Hexadimetrina/farmacologia , Humanos , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo , Transdução Genética , Transgenes/genética , Células Tumorais Cultivadas , Células Vero
9.
Hum Gene Ther ; 9(18): 2787-94, 1998 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-9874276

RESUMO

Herpes simplex virus type 1 (HSV-1)-based amplicon vectors contain only approximately 1% of the 152-kb HSV-1 genome, and consequently, replication and packaging into virions depends on helper functions. These helper functions have been provided conventionally by a helper virus, usually a replication-defective mutant of HSV-1, or more recently, by a set of five cosmids that overlap and represent the genome of HSV-1 deleted for DNA cleavage/packaging signals (pac). In the absence of pac signals, potential HSV-1 genomes that are reconstituted from the cosmids via homologous recombination are not packageable. The resulting amplicon stocks are, therefore, virtually free of contaminating helper virus. To simplify this packing system, the HSV-1 genome was cloned and maintained stably as a single-copy, F plasmid-based bacterial artificial chromosome in E. coli. Such a plasmid containing the HSV-1 genome deleted for the pac signals (fHSV delta pac) did not generate replication-competent progeny virus on transfection into mammalian cells, but rather, it was able to support the packaging of cotransfected amplicon DNA that contained a functional pac signal. The resulting amplicon vector stocks had titers of up to 10(7) transducing units per milliliter of culture medium and efficiently transduced neural cells in the rat brain, as well as hepatocytes in the rat. The capacity of generating infectious and replication-competent HSV-1 progeny following transfection into mammalian cells was restored after insertion of a pac signal into fHSV delta pac.


Assuntos
Cromossomos Bacterianos/genética , Escherichia coli/genética , Vetores Genéticos , Herpesvirus Humano 1/genética , Montagem de Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , Cosmídeos , Efeito Citopatogênico Viral , DNA Viral/genética , Técnicas de Transferência de Genes , Herpesvirus Humano 1/fisiologia , Hipocampo/virologia , Humanos , Fígado/virologia , Plasmídeos/genética , Ratos , Transfecção , Células Vero , Vírion/genética
10.
Neoplasia ; 1(5): 402-16, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10933055

RESUMO

Many properties of HSV-1 are especially suitable for using this virus as a vector to treat diseases affecting the central nervous system (CNS), such as Parkinson's disease or malignant gliomas. These advantageous properties include natural neurotropism, high transduction efficiency, large transgene capacity, and the ability of entering a latent state in neurons. Selective oncolysis in combination with modulation of the immune response mediated by replication-conditional HSV-1 vectors appears to be a highly promising approach in the battle against malignant glioma. Helper virus-free HSV/AAV hybrid amplicon vectors have great promise in mediating long-term gene expression in the PNS and CNS for the treatment of various neurodegenerative disorders or chronic pain. Current research focuses on the design of HSV-1-derived vectors which are targeted to certain cell types and support transcriptionally regulatable transgene expression. Here, we review the recent developments on HSV-1-based vector systems and their applications in experimental and clinical gene therapy protocols.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Doenças do Sistema Nervoso/terapia , Genes Virais , Humanos , Modelos Genéticos , Neurônios/metabolismo
11.
Neoplasia ; 1(5): 387-401, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10933054

RESUMO

The design of effective gene therapy strategies for brain tumors and other neurological disorders relies on the understanding of genetic and pathophysiological alterations associated with the disease, on the biological characteristics of the target tissue, and on the development of safe vectors and expression systems to achieve efficient, targeted and regulated, therapeutic gene expression. The herpes simplex virus type 1 (HSV-1) virion is one of the most efficient of all current gene transfer vehicles with regard to nuclear gene delivery in central nervous system-derived cells including brain tumors. HSV-1-related research over the past decades has provided excellent insight into the structure and function of this virus, which, in turn, facilitated the design of innovative vector systems. Here, we review aspects of HSV-1 structure, replication and pathogenesis, which are relevant for the engineering of HSV-1-based vectors.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Doenças do Sistema Nervoso/terapia , Técnicas de Transferência de Genes , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/ultraestrutura , Humanos , Modelos Biológicos
12.
Neuroreport ; 8(17): 3801-8, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9427374

RESUMO

Green fluorescent protein (GFP) is an effective marker for retrovirus and herpes virus vector-mediated gene transfer into various central nervous system-derived cells, both proliferative and non-proliferative, in culture and in vivo. Retrovirus vectors were used to stably transduce several rat and human glioma lines, and a multipotent mouse neural progenitor line in culture. Implantation of selected pools of transduced glioma cells into rodent brain allowed clear visualization of the tumor and the invading tumor edge. Helper virus-free HSV-1 amplicon vectors successfully transferred gfp into non-dividing primary neural cells in culture and in the rat brain. This study describes the versatility of GFP for: (i) labelling of glioma cells in experimental brain tumor models and neural progenitor cells by retrovirus vectors, and (ii) efficient, non-toxic delivery of genes to post mitotic cells of the nervous system using helper-virus free HSV-1 amplicon vectors.


Assuntos
Neoplasias Encefálicas/patologia , Técnicas de Transferência de Genes , Glioma/patologia , Herpesvirus Humano 1/genética , Proteínas Luminescentes/biossíntese , Animais , Capsídeo/análise , Capsídeo/biossíntese , Núcleo Celular/ultraestrutura , Células Cultivadas , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/análise , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Neurônios/citologia , Ratos , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/biossíntese , Células-Tronco , Células Tumorais Cultivadas
13.
Vet Microbiol ; 86(1-2): 103-13, 2002 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-11888694

RESUMO

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.


Assuntos
Doenças do Gato/virologia , Terapia Genética/veterinária , Herpes Simples/veterinária , Herpesvirus Humano 1/genética , Interleucina-10/genética , Interleucina-6/genética , Animais , Doenças do Gato/terapia , Gatos , Chlorocebus aethiops , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Herpes Simples/terapia , Herpesvirus Humano 1/química , Interferon gama/biossíntese , Interferon gama/genética , Interleucina-10/biossíntese , Interleucina-12/biossíntese , Interleucina-12/genética , Interleucina-6/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Vero
14.
Vet Microbiol ; 86(1-2): 165-74, 2002 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-11888699

RESUMO

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.


Assuntos
Terapia Genética/veterinária , Herpesvirus Bovino 1/genética , Proteínas Virais/genética , Animais , Bovinos , Chlorocebus aethiops , Efeito Citopatogênico Viral/genética , Citometria de Fluxo/veterinária , Terapia Genética/métodos , Vetores Genéticos , Proteínas de Fluorescência Verde , Herpesvirus Bovino 1/química , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Proteínas Luminescentes/química , Monócitos , Plasmídeos/genética , Transdução Genética/métodos , Células Vero , Proteínas Virais/química
15.
Adv Exp Med Biol ; 451: 393-403, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-10026902

RESUMO

Quantity and distribution of transgene-expressing tumor cells are central issues in cancer gene therapy. These are critical for the efficiency of tumor killing and for the bystander effect. In an attempt to combine the advantages of a potent bioactivating "suicide" gene with a marker gene for living cells, cDNA encoding cytochrome CYP4B1 was fused to the green fluorescent protein (GFP) cDNA. The resulting chimeric fusion protein, 4B1EGFP, was expressed in rodent and human glioma cell lines in culture. The ability of this recombinant enzyme to destroy tumor cells by converting the prodrug 4-ipomeanol (4-IM) into alkylating metabolites was evaluated in comparison with the cytotoxicity of the native CYP4B1 enzyme. The most sensitive 4B1EGFP-expressing glioma cell clone had a LD50 of 0.75 microgram/ml for 4-IM, as compared to a 4-IM LD50 of 0.5 microgram/ml in glioma cells expressing the native CYP4B1. A strong bystander effect mediated by cell-to-cell contact was present in the 4B1EGFP clones, allowing for more than 50% bystander kill at a ratio of expressing to non-expressing cells of 1:100. A herpes-simplex amplicon (pHSVPrPUC delta Hind) was constructed with the 4B1EGFP fusion protein, and recombinant helper-free HSV particles were packaged in Vero cells. Fisher 344 rats were inoculated with 4 x 10(5) 9L tumor cells to produce epidural tumor. Recombinant HSV particles were injected into the tumor at a dose of 1 x 10(7) pfu. Tumor was resected in living anesthetized animals 24, 48, and 72 hours after virus injection, and cryostat sections were evaluated by fluorescent microscopy. HSV-mediated delivery of the fusion protein to tumor cells was successfully demonstrated. In conclusion, the chimeric fusion protein 4B1EGFP retains essentially all features of the native CYP4B1 enzyme, and, moreover, offers advantages in terms of gene transfer visualization, which may lead to improvement of gene transfer strategies.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/genética , Terapia Genética , Glioblastoma/terapia , Gliossarcoma/terapia , Pró-Fármacos/farmacocinética , Terpenos/farmacocinética , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Apoptose , Sequência de Bases , Divisão Celular , Sobrevivência Celular/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/análise , Sistema Enzimático do Citocromo P-450/biossíntese , Fragmentação do DNA , DNA Complementar , Glioblastoma/patologia , Gliossarcoma/patologia , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Dados de Sequência Molecular , Pró-Fármacos/toxicidade , Ratos , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/biossíntese , Mapeamento por Restrição , Terpenos/toxicidade , Células Tumorais Cultivadas
16.
Gene Ther ; 14(15): 1143-51, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17495946

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Glioma/terapia , Herpesvirus Humano 1/genética , Rad51 Recombinase/antagonistas & inibidores , Animais , Apoptose , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Expressão Gênica , Inativação Gênica , Vetores Genéticos/genética , Glioblastoma/terapia , Glioma/patologia , Glioma/radioterapia , Humanos , Injeções Intralesionais , Camundongos , Camundongos Nus , Interferência de RNA , RNA Interferente Pequeno/genética , Rad51 Recombinase/genética , Tolerância a Radiação/fisiologia , Resultado do Tratamento
17.
Gene Ther ; 12(16): 1283-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15889134

RESUMO

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.


Assuntos
Proteínas do Olho/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Herpesvirus Humano 1/genética , Retina/metabolismo , Transdução Genética/métodos , Animais , Proteínas de Transporte , Citomegalovirus/genética , Proteínas do Olho/análise , Proteínas do Olho/metabolismo , Expressão Gênica , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Injeções , Microscopia de Fluorescência , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Doenças Retinianas/terapia , Fatores de Tempo , cis-trans-Isomerases
18.
Gene Ther ; 12(23): 1707-17, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16034459

RESUMO

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.


Assuntos
Vacinas Anticâncer/genética , Terapia Genética/métodos , Herpesvirus Humano 1/genética , Imunoterapia/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Ligante CD27 , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Genes MHC Classe I , Engenharia Genética , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Transdução Genética/métodos , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/imunologia
19.
Gene Ther ; 12(7): 588-96, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15674397

RESUMO

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.


Assuntos
Neoplasias Encefálicas/virologia , Vetores Genéticos , Herpesvirus Humano 1/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Moléculas de Adesão Celular/metabolismo , Proliferação de Células , Expressão Gênica , Herpesvirus Humano 1/patogenicidade , Humanos , Nectinas , Proteínas de Neoplasias/metabolismo , Receptores Virais/metabolismo , Fatores de Tempo , Transdução Genética , Células Tumorais Cultivadas
20.
J Virol ; 68(12): 8082-8, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7966598

RESUMO

We have recently reported immediate-early (IE) transcription over covalently joined genome ends of bovine herpesvirus 1 (BHV-1). A spliced 1.5-kb IE RNA (IER1.5) is coterminal with an unspliced 1.1-kb late RNA (LR1.1) which is transcribed from the left end of the genome. Sequence analysis reveals an open reading frame common to IER1.5 and LR1.1 predicted to encode the 247-amino-acid circ polypeptide. This paper reports on the identification of circ as a protein. Using a rabbit antiserum raised against a synthetic oligopeptide representing the carboxy terminus of the predicted circ polypeptide for Western blot (immunoblot) analyses and immunofluorescence assays, we identified a 34-kDa virion-associated protein which accumulated in the cytoplasm of infected cells. To confirm that LR1.1 indeed encoded the 34-kDa polypeptide, we inserted a DNA fragment containing circ coding sequences into the Autographa californica baculovirus genome. A group of recombinant polypeptides with sizes of 32, 34, and 35 kDa were identified by their reactivity with the antipeptide serum. Chicken egg yolk antibodies raised against total proteins of insect cells infected with the recombinant baculovirus identified the 34-kDa circ protein specified by BHV-1. The recombinant circ polypeptides and the circ protein specified by BHV-1 were both myristylated, as determined by radiolabeling with [3H]myristic acid. It was noted that the circ gene could be deleted from the BHV-1 genome without impairing virus replication in cell culture.


Assuntos
Genes Virais , Herpesvirus Bovino 1/fisiologia , Proteínas Virais/biossíntese , Vírion/fisiologia , Replicação Viral , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Deleção de Genes , Genoma Viral , Herpesvirus Bovino 1/genética , Soros Imunes , Rim , Dados de Sequência Molecular , Ácido Mirístico , Ácidos Mirísticos/metabolismo , Splicing de RNA , RNA Viral/biossíntese , RNA Viral/metabolismo , Proteínas Virais/análise , Vírion/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA