Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cancer Res ; 61(7): 2983-95, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11306477

RESUMEN

To evaluate the efficiency of gene delivery in gene therapy strategies for malignant brain tumors, it is important to determine the distribution and magnitude of transgene expression in target tumor cells over time. Here, we assess the time- and vector dose-dependent kinetics of recombinant herpes simplex virus (HSV)-1 vector-mediated gene expression and vector replication in culture and in vivo by a recently developed radiotracer method for noninvasive imaging of gene expression (J. G. Tjuvajev et al., Cancer Res., 55: 6126-6132, 1995). The kinetics of viral infection of rat 9L gliosarcoma cells by the replication-conditional HSV-1 vector, hrR3, was studied by measuring the accumulation rate of 2-[14C]-fluoro-5-iodo-1-beta-D-arabinofuranosyl-uracil (FIAU), a selective substrate for viral thymidine kinase (TK). The level of viral TK activity in 9L cells was monitored by the radiotracer assay to assess various vector doses and infection times, allowing vector replication and spread. In parallel, viral yields and levels of Escherichia coli beta-galactosidase activity were assessed quantitatively. To study vector replication, spread and HSV-1-tk and lacZ gene coexpression in vivo, first- or second-generation recombinant HSV-1 vectors (hrR3 or MGH-1) were injected into s.c. growing rat 9L or human U87 deltaEGFR gliomas in nude rats at various times (8 h to 8 days) and at various vector doses [1 x 10(6) to 2 x 10(9) plaque-forming units (PFUs)] prior to imaging. For noninvasive assessment of HSV-1-tk gene expression (124I-labeled FIAU % dose/g), 0.15 mCi of 124I-labeled FIAU was injected i.v. 8 h after the last vector administration, and FIAU positron emission tomography (PET) was performed 48 h later. For the assessment of HSV-1-tk and lacZ gene coexpression, 0.2 mCi of 131I-labeled FIAU was injected i.v. 24 h after the last vector administration. Forty-eight h later, animals were killed, and tumors were dissected for quantitative autoradiographical and histochemical assessment of regional distribution of radioactivity (TK expression measured as 131I-labeled FIAU % dose/g) and coexpressed lacZ gene activity. The rates of FIAU accumulation (Ki) in hrR3-infected 9L cells in culture, which reflect the levels of HSV-1-tk gene expression, ranged between 0.12 and 3.4 ml/g/min. They increased in a vector dose- and infection time-dependent manner and correlated with the virus yield (PFUs/ml), where the PFUs:Ki ratios remained relatively constant over time. Moreover, a linear relationship was observed between lacZ gene expression and FIAU accumulation 5-40 h after infection of 9L cells with a multiplicity of infection of 1.5. At later times (> 52 h postinjection), high vector doses (multiplicity of infection, 1.5) led to a decrease of FIAU accumulation rates, viral yield, and cell pellet weights, indicating vector-mediated cell toxicity. Various levels of HSV-1-tk gene expression could be assessed by FIAU-PET after in vivo infection of s.c. tumors. The levels of FIAU accumulation were comparatively low (approximately ranging from 0.00013 to 0.003% injected dose/g) and were spatially localized; this may reflect viral-induced cytolysis of infected tumor cells and limited lateral spread of the virus. Image coregistration of tumor histology, HSV-1-tk related radioactivity (assessed by autoradiography), and lacZ gene expression (assessed by beta-galactosidase staining) demonstrated a characteristic pattern of gene expression around the injection sites. A rim of lacZ gene expression immediately adjacent to necrotic tumor areas was observed, and this zone was surrounded by a narrow band of HSV-1-tk-related radioactivity, primarily in viable-appearing tumor tissue. These results demonstrate that recombinant HSV-1 vector-mediated HSV-1-tk gene expression can be monitored noninvasively by PET, where the areas of FIAU-derived radioactivity identify the viable portion of infected tumor tissue that retains FIAU accumulation ability, and that the accumulation rate of FIAU in culture, Ki, reflects the number of HSV-1 viral particles in the infected tumor cell population [4.1 +/- 0.6 x 10(6) PFUs/Ki unit (PFUs divided by ml/min/g)]. Moreover, time-dependent and spatial relationships of HSV-1-tk and lacZ gene coexpression in culture and in vivo indicate the potential for indirect in vivo imaging of therapeutic gene expression in tumor tissue infected with any recombinant HSV-1 vector where a therapeutic gene is substituted for the lacZ gene.


Asunto(s)
Arabinofuranosil Uracilo/análogos & derivados , Regulación Viral de la Expresión Génica , Herpesvirus Humano 1/fisiología , Transgenes , Animales , Arabinofuranosil Uracilo/farmacocinética , Autorradiografía , Chlorocebus aethiops , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glioma/genética , Gliosarcoma/genética , Herpesvirus Humano 1/genética , Humanos , Radioisótopos de Yodo , Operón Lac/genética , Ratones , Ratones Desnudos , Mutación , Ratas , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética , Tomografía Computarizada de Emisión , Células Vero , Replicación Viral
2.
Hum Gene Ther ; 7(3): 291-300, 1996 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-8835217

RESUMEN

The present study investigated the ability of a recombinant herpes simplex virus type 1 (HSV) vector to deliver genes into disseminated brain tumor foci through intrathecal injection of the vector. The animal model was designed to simulate brain tumors with cerebrospinal fluid (CSF) metastases, which are found especially in the pediatric population. 9L gliosarcoma cells were injected both into the right frontal lobe and in through the cisterna magna of adult rats. The HSV vector, hrR3, was inoculated intrathecally 5 days later. This vector is defective in the gene for ribonucleotide reductase, and, therefore, replicates preferentially in dividing cells; it retains an intact HSV-thymidine kinase gene (HSV-tk). Two days after injection of the vector, immunohistochemical staining for HSV thymidine kinase (HSV-TK) revealed expression in frontal tumors, as well as in leptomeningeal tumor foci along the entire neuroaxis. HSV-TK-immunopositive cells were most frequent in small tumors contacting the CSF pathways. Frontal lobe tumors showed the highest density of HSV-TK-immunopositive cells around their periphery with little expression in central parts. Some paraventricular neurons temporarily showed HSV-TK-immunolabeling at this early time point. The number of HSV-TK-immunopositive tumor cells markedly decreased 5 days after injection of the HSV vector. In all animals, some toxicity was observed in the first 2-4 days after virus injection with extensive leptomeningeal inflammation. In conclusion, intrathecal application of HSV vectors can mediate widespread transfer of the therapeutic HSV-tk gene into disseminated tumors throughout the brain and CSF pathways. Although there was marked toxicity associated with intrathecal injection of this vector, this mode of gene delivery offers a promising approach for treatment of CSF-metastases in conjunction with development of less toxic vectors.


Asunto(s)
Neoplasias del Sistema Nervioso Central/terapia , Cisterna Magna , Lóbulo Frontal , Genes Reporteros , Vectores Genéticos/genética , Gliosarcoma/secundario , Neoplasias Meníngeas/secundario , Simplexvirus/genética , Transfección , Animales , Neoplasias del Sistema Nervioso Central/patología , Genes Sintéticos , Gliosarcoma/patología , Gliosarcoma/terapia , Inyecciones Espinales , Masculino , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/terapia , Metástasis de la Neoplasia , Trasplante de Neoplasias , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes/análisis , Simplexvirus/patogenicidad , Simplexvirus/fisiología , Espacio Subaracnoideo , Timidina Quinasa/análisis , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética , Replicación Viral , beta-Galactosidasa/análisis , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
3.
Hum Gene Ther ; 7(16): 1989-94, 1996 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-8930659

RESUMEN

Brain tumors that have disseminated into cerebrospinal fluid (CSF) pathways are an unresolved therapeutic problem, especially in pediatric neurooncology. Here a gene therapy approach using the herpes simplex virus type 1 thymidine kinase (HSV-TK)/ganciclovir (GCV) paradigm was tested using an HSV vector in a rodent model of disseminated central nervous system tumors. 9L-gliosarcoma cells were implanted simultaneously into the brain and the CSF of syngeneic rats. Five days later, resulting intracerebral and leptomeningeal tumors were treated by intrathecal injection of a replication-conditional HSV vector. This vector was defective for the ribonucleotide reductase gene, but contained an intact HSV-tk gene. Systemic GCV treatment was started 2 days after vector application and continued for 14 days. Tumor-free, long-term survival (LTS) was achieved in 90% of the animals treated with this combined therapeutic approach, whereas only 30% LTS was found in animals that had received the vector alone and 10% LTS in untreated animals. This therapeutic response probably involves oncolytic, on-site replication of the vector, activation of GCV by a HSV-TK, and a strong immune response both to the vector and to 9L cells. Apparent vector-related mortality was observed in 20% of animals without subsequent GCV therapy, but no vector-related mortality was found when the animals were treated with GCV after vector application. Given the successful outcome of this experimental treatment and the apparent potential of GCV to control HSV-related toxicity, intrathecal application of HSV vectors combined with GCV treatment may be a promising approach for treatment of disseminated brain tumors.


Asunto(s)
Antivirales/farmacología , Neoplasias Encefálicas/terapia , Ganciclovir/farmacología , Vectores Genéticos/fisiología , Gliosarcoma/terapia , Herpesvirus Humano 1/fisiología , Animales , Modelos Animales de Enfermedad , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/genética , Humanos , Inyecciones Espinales , Neoplasias Experimentales , Ratas , Timidina Quinasa/genética , Factores de Tiempo , Células Tumorales Cultivadas
4.
Hum Gene Ther ; 7(16): 2003-13, 1996 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-8930661

RESUMEN

Recombinant and amplicon vectors derived from herpes simplex virus type 1 (HSV-1) have proven to be an efficient means of gene delivery to cells in culture and in vivo. In this study, a system was developed to make propagation of the amplicon vector and helper virus mutually dependent on each other, in a "piggyback' fashion. This combined system supports maintenance and enrichment of the amplicon vector when propagating stocks, while allowing the helper virus to serve as a recombinant vector in its own right. Amplicons bearing a gene essential for HSV-1 replication, IE3, as well as the Escherichia coli lacZ marker gene, were propagated using a mutant virus (d120) deleted in the same essential gene. Vector stocks could be propagated in Vero cells and other cultured cells not transfected with the IE3 gene with markedly delayed cytopathic effects, as compared to wild-type virus. Relatively high titers of amplicon vectors (6 x 10(7) infectious units/ml) were achieved with this piggyback system in Vero cells, with an apparent ratio of amplicon vector: helper virus of up of 5:1 under some conditions; however, recombinant wild-type virus was also generated. Injection of these stocks into experimental gliomas in rodent brain revealed gene delivery to tumor cells mediated by both amplicon vectors (lacZ) and helper virus (HSV-thymidine kinase), with no apparent neuropathology of normal brain. This basic piggyback vector model is amenable to modifications to promote conditional propagation of vectors in vivo and to allow incorporation of multiple transgene elements into both the amplicon and recombinant helper virus vectors.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Virus Helper/genética , Herpesvirus Humano 1/genética , Animales , Southern Blotting , Línea Celular , Chlorocebus aethiops , Humanos , Plásmidos , Células Vero
5.
Hum Gene Ther ; 8(3): 331-9, 1997 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-9048200

RESUMEN

The therapeutic use of neurotrophic factors for neurodegenerative diseases is promising, however, optimal methods for continuous delivery of these substances to the human central nervous system (CNS) remains problematic. One approach would be to graft genetically engineered human cells that continuously secrete high levels of a biologically produced and processed neurotrophic factor. This ex vivo gene therapy approach has worked well in animal models of neurodegenerative diseases using a variety of nonneuronal cell types to deliver the transgene. In our studies, we have been investigating the potential of astrocytes, a cell type normally present in the CNS, as a vehicle for ex vivo gene therapy. Here, we demonstrate that astrocytes in the human fetal cortex can be isolated and efficiently infected with an amphotropic retrovirus harboring mouse beta-nerve growth factor (NGF). These transduced astrocytes express high levels of NGF mRNA and secrete bioactive NGF protein as demonstrated by stimulation of neurite outgrowth from adrenal chromaffin cells. NGF ELISA showed that these astrocytes secrete NGF protein at a rate of 41 ng/day per 10(5) cells after 2 weeks in vitro, whereas NGF is undetectable in medium conditioned by normal astrocytes. These data suggest that human fetal astrocytes can be used for delivering biologically produced neurotrophic factors to the human CNS.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/trasplante , Trasplante de Células/métodos , Corteza Cerebral/citología , Terapia Genética/métodos , Factores de Crecimiento Nervioso/biosíntesis , Línea Celular , Células Cromafines , Feto , Vectores Genéticos/genética , Humanos , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/uso terapéutico , Retroviridae , Transfección/genética , Transfección/métodos
6.
Hum Gene Ther ; 8(3): 359-70, 1997 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-9048203

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes Virales , Vectores Genéticos , Glioma/genética , Herpesvirus Humano 1/genética , Proteínas Virales/genética , Animales , Western Blotting , Chlorocebus aethiops , Proteínas de Unión al ADN/biosíntesis , Dependovirus/genética , Expresión Génica , Técnicas de Transferencia de Gen , Genes Reporteros , Glioma/terapia , Glioma/virología , Humanos , Reacción en Cadena de la Polimerasa , Células Vero , Proteínas Virales/biosíntesis
7.
Hum Gene Ther ; 8(17): 2057-68, 1997 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-9414254

RESUMEN

A second-generation replication-conditional herpes simplex virus type 1 (HSV) vector defective for both ribonucleotide reductase (RR) and the neurovirulence factor gamma34.5 was generated and tested for therapeutic safety and efficiency in two different experimental brain tumor models. In culture, cytotoxic activity of this double mutant HSV vector, MGH-1, for 9L gliosarcoma cells was similar to that of the HSV mutant, R3616, which is defective only for gamma34.5, but was significantly weaker than that of the HSV mutant hrR3, which is defective only for RR. The diminished tumoricidal effect of the gamma34.5 mutants could be accounted for by their reduced ability to replicate in 9L cells. The MGH-1 vector did not achieve significant prolongation of survival in vivo in the syngeneic 9L rat gliosarcoma model for either single brain tumor focus or multiple intracerebral and leptomeningeal tumors, when the vector was applied intratumorally or intrathecally, respectively, and with or without subsequent ganciclovir (GCV) treatment. In identical 9L brain tumor models with single and multiple foci, application of hrR3 with or without GCV was previously shown to result in marked long-term survival. Contrary to the findings with intrathecal injection of hrR3, no vector-related mortality was observed in any animals treated with MGH-1. Thus, in these rat brain tumor models, the double mutant, replication-conditional HSV vector MGH-1 showed a higher therapeutic safety than the RR-minus vector, hrR3, but had clearly decreased therapeutic efficiency compared to hrR3. The development of new HSV vectors for brain tumor gene therapy will require a balance between maximizing therapeutic efficacy and minimizing toxicity to the brain. Standardized application in brain tumor models as presented here will help to screen new HSV vectors for these requirements.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética , Vectores Genéticos , Gliosarcoma/terapia , Herpesvirus Humano 1/genética , Ribonucleótido Reductasas/genética , Proteínas Virales/genética , Animales , Chlorocebus aethiops , Terapia Combinada , Modelos Animales de Enfermedad , Ganciclovir/uso terapéutico , Eliminación de Gen , Vectores Genéticos/toxicidad , Gliosarcoma/secundario , Humanos , Masculino , Neoplasias Meníngeas/secundario , Ratas , Ratas Endogámicas F344 , Ribonucleótido Reductasas/metabolismo , Células Tumorales Cultivadas , Células Vero , Proteínas Virales/metabolismo , Replicación Viral
8.
Brain Pathol ; 5(4): 345-81, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8974620

RESUMEN

Gene therapy has opened new doors for treatment of neoplastic diseases. This new approach seems very attractive, especially for glioblastomas, since treatment of these brain tumors has failed using conventional therapy regimens. Many different modes of gene therapy for brain tumors have been tested in culture and in vivo. Many of these approaches are based on previously established anti-neoplastic principles, like prodrug activating enzymes, inhibition of tumor neovascularization, and enhancement of the normally weak anti-tumor immune response. Delivery of genes to tumor cells has been mediated by a number of viral and synthetic vectors. The most widely used paradigm is based on the activation of ganciclovir to a cytotoxic compound by a viral enzyme, thymidine kinase, which is expressed by tumor cells, after the gene has been introduced by a retroviral vector. This paradigm has proven to be a potent therapy with minimal side effects in several rodent brain tumor models, and has proceeded to phase 1 clinical trials. In this review, current gene therapy strategies and vector systems for treatment of brain tumors will be described and discussed in light of further developments needed to make this new treatment modality clinically efficacious.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética , Formación de Anticuerpos , Ensayos Clínicos como Asunto , Terapia Genética/métodos , Vectores Genéticos , Humanos , Neovascularización Patológica , Transgenes , Virus/genética
9.
Neuroreport ; 3(6): 469-72, 1992 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-1391749

RESUMEN

Hydrogen peroxide (H2O2) is a type of active oxygen species produced mainly in blood by inflammation, ischemia or anoxia. Treatment of rat neonatal cortical astrocytes in culture with 0.2-1.0 mM H2O2 which is lethal for hippocampal neurons, increases nerve growth factor (NGF) and basic fibroblast growth factor (bFGF) mRNA content in a time dependent manner. H2O2 also increases c-fos mRNA expression, which is probably involved in the gene regulation of both NGF and bFGF. Maximal induction was reached after 6 h of incubation (5.7-fold increase in NGF and 2.4-fold induction of bFGF mRNA). Hydrogen peroxide induced bFGF and NGF gene expression suggests that neurotrophic factors in astrocytes could be induced by lesion, consistent with their protective function in the CNS.


Asunto(s)
Astrocitos/fisiología , Factor 2 de Crecimiento de Fibroblastos/genética , Peróxido de Hidrógeno/farmacología , Factores de Crecimiento Nervioso/genética , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Northern Blotting , Encéfalo/fisiología , Células Cultivadas , Radicales Libres , Expresión Génica/efectos de los fármacos , Genes fos , Cinética , ARN Mensajero/biosíntesis , Ratas , Factores de Tiempo
10.
Neuroreport ; 3(12): 1077-80, 1992 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1493220

RESUMEN

Basic FGF mRNA induction by bFGF was investigated in cell cultures from rat brain, i.e. postnatal day 2 cortex and embryonic day 18 hippocampus. In situ hybridization shows that after bFGF treatment (10(-10) M) for 14 h neurones and glial cells show a remarkable increase in bFGF mRNA production. Incubation of astrocytes with antisense bFGF phosphorothioate oligodeoxynucleotides (bFGF-PTOs) resulted in an inhibition of both bFGF induced and serum induced proliferation. The results indicate that bFGF is capable of inducing its own mRNA production. This induction, i.e. new synthesis of bFGF mRNA, seems to be essential for the mitogenic effect of both bFGF and serum components.


Asunto(s)
Astrocitos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Digoxigenina , Factor 2 de Crecimiento de Fibroblastos/genética , Hipocampo/efectos de los fármacos , Hibridación in Situ , Sondas ARN , ARN sin Sentido/metabolismo , Ratas
11.
Neuroreport ; 6(4): 669-72, 1995 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-7605924

RESUMEN

The neuroprotective effect of nerve growth factor (NGF) on the pyramidal cells in the vulnerable CA1-CA2 sectors of the hippocampus was investigated in a rat model of transient forebrain ischemia. A genetically modified fibroblast line that secretes high levels of NGF was implanted 7 days before induction of ischemia between the hippocampal CA1-CA2 subfields in the right hemisphere. Rats were then subjected to 10 min of cerebral ischemia in a four vessel occlusion model. Morphological changes in the CA1 and CA2 subfields were evaluated 7 days after ischemia. Animals in the NGF-protected group had significantly higher numbers of normal appearing neurons in the right CA1 and CA2 regions, compared with their non-implanted left hemispheres, to non-implanted animals or to animals implanted with non-modified cells. The data confirmed that NGF can protect CA1-CA2 hippocampal neurons from ischemic damage by implantation of genetically engineered cells producing NGF.


Asunto(s)
Fibroblastos/metabolismo , Terapia Genética , Ataque Isquémico Transitorio/tratamiento farmacológico , Factores de Crecimiento Nervioso/farmacología , Prosencéfalo/irrigación sanguínea , Animales , Línea Celular , Fibroblastos/trasplante , Hipocampo/citología , Hipocampo/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar
12.
Neuroreport ; 8(17): 3801-8, 1997 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9427374

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/patología , Técnicas de Transferencia de Gen , Glioma/patología , Herpesvirus Humano 1/genética , Proteínas Luminiscentes/biosíntesis , Animales , Cápside/análisis , Cápside/biosíntesis , Núcleo Celular/ultraestructura , Células Cultivadas , Genes Reporteros , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/análisis , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Neuronas/citología , Ratas , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/biosíntesis , Células Madre , Células Tumorales Cultivadas
13.
Neurosci Lett ; 153(1): 111-4, 1993 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-8099723

RESUMEN

The effect of glutamate on primary cultures of rat cortical astrocytes was studied using Northern blot hybridization. Incubation with glutamate (100 microM, 15 min) induced nerve growth factor (NGF), basic fibroblast growth factor (bFGF), FGF receptor (FGF-R1) and proto-oncogene c-fos gene expression in a time dependent manner. Maximal induction of NGF, bFGF and FGF-R1 mRNA was reached after 4 h of incubation (7.2-fold induction of NGF, 3-fold increase in bFGF and 3.6-fold induction of FGF-R1 mRNA). The induction kinetics of NGF, bFGF and FGF-R1 mRNA are similar. The rapid (1 h) 77-fold induction of the c-fos transcript precedes the induction of the other genes tested.


Asunto(s)
Astrocitos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Glutamatos/farmacología , Factores de Crecimiento Nervioso/genética , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Animales , Northern Blotting , Células Cultivadas , Expresión Génica , Ácido Glutámico , Ratas
14.
Teratology ; 33(2): 239-41, 1986 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-3738819

RESUMEN

Syngeneic spleen cell immunization of C57BL/6J females prior to syngeneic matings resulted in significant perinatal and postnatal mortality and increased abnormality among progeny. Thus female recipients of transplants and transfusions from major histocompatibility complex (MHC) compatible donors might effect some risk to their progeny.


Asunto(s)
Muerte Fetal/etiología , Antígeno H-Y/administración & dosificación , Bazo/inmunología , Animales , Femenino , Cabeza/anomalías , Humanos , Inmunización , Masculino , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Embarazo , Factores Sexuales , Bazo/trasplante , Teratógenos , Trasplante Isogénico
15.
J Gene Med ; 1(3): 176-85, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10738566

RESUMEN

BACKGROUND: The versatility of HSV-1 vectors includes large transgene capacity, selective replication of mutants in dividing cells, and availability of recombinant virus (RV) and plasmid-derived (amplicon) vectors, which can be propagated in a co-dependent, 'piggyback', manner. METHODS: A replication-defective piggyback vector system was generated in which the amplicon carries either of two genes essential for virus replication, IE2 (ICP27) or IE3 (ICP4), as well as lacZ; the RV is deleted in both these genes, and vector stocks are propagated in cells transfected with one of the complementary genes. In the replication-competent system, the amplicon carries the IE2 and lacZ; the RV had a large deletion in the IE2; and stocks are propagated in untransfected cells. Titers over successive passages, recombination between amplicon and RV, and the structural integrity of vector genomes were evaluated. The replication-competent system was tested for therapeutic efficacy in subcutaneous 9L gliosarcoma tumors in nude mice with activation of ganciclovir via the viral HSV-thymidine kinase gene. RESULTS: Both systems generated high titer amplicon vectors (about 10(7) tu/ml) and amplicon:RV ratios (0.6-3.0). No replication-competent RV was generated in either system. The replication-defective system showed low toxicity and increased packaging efficiency of amplicon vectors, as compared to single mutant RV helper virus. The replication-competent system allowed co-propagation of amplicon and RV; injection into tumors followed by ganciclovir treatment inhibited tumor growth without systemic toxicity. CONCLUSION: New replication-defective and replication-competent piggyback HSV, vector systems allow gene delivery via amplicon vectors with reduced toxicity and co-propagation of both RV and amplicon vectors in target cells, with effective tumor therapy via focal virus replication and pro-drug activation.


Asunto(s)
Terapia Genética , Vectores Genéticos , Gliosarcoma/terapia , Herpesvirus Humano 1/genética , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Cricetinae , Virus Defectuosos/genética , Virus Defectuosos/fisiología , Genes Inmediatos-Precoces , Virus Helper/genética , Virus Helper/fisiología , Herpesvirus Humano 1/fisiología , Operón Lac , Masculino , Ratones , Ratones Desnudos , Plásmidos/genética , Ratas , Recombinación Genética , Células Vero , Replicación Viral
16.
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.
Gene Ther ; 5(6): 809-19, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9747461

RESUMEN

The influence of pre-existing anti-herpes simplex type 1 (HSV-1) immunity on HSV-1 vector-mediated gene transfer to glioma cells was analyzed in this gene marking study using intracranial D74 gliomas in syngeneic Fischer rats. The HSV-1 mutant virus used, hrR3, is defective in ribonucleotide reductase and bears the marker genes E. coli lacZ and HSV-1 thymidine kinase (HSVtk). Initial marker gene expression in tumors 12 h after direct virus injection was reduced in immunized animals to about 15% of that in nonimmunized animals. Marker gene expression in both sets stayed at initial levels for 2 days after intratumoral injection and declined markedly on day 5. Inflammatory infiltrates in the tumor were more prominent in HSV-1-immunized, as compared with nonimmunized animals, at 12 and 24 h, but appeared similar at 2-5 days after injection. By day 10, the immune reaction had subsided in immunized animals and macrophages remained only in nonimmunized animals. In conclusion, gene transfer to brain tumors using a HSV-1 vector was greatly reduced, but not completely abolished, under pre-immunization conditions. Pre-existing antibodies to HSV-1 may also serve a positive role in providing an increased margin of safety in intracranial application of HSV-1 vectors by limiting spread of the virus within the brain and to other tissues.


Asunto(s)
Neoplasias Encefálicas/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Glioma/terapia , Simplexvirus/inmunología , Animales , Neoplasias Encefálicas/inmunología , Expresión Génica , Marcadores Genéticos , Glioma/inmunología , Inmunización , Ratas , Ratas Endogámicas F344 , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA