Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 22(11): 893-900, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26090874

RESUMO

The soluble vascular endothelial growth factor (VEGF) receptor 1 (sFLT1) has been tested in both animals and humans for anti-angiogenic therapies, for example, age-related macular degeneration. We hypothesized that adeno-associated viral vector (AAV)-mediated sFLT1 expression could be used to inhibit abnormal brain angiogenesis. We tested the anti-angiogenic effect of sFLT1 and the feasibility of using AAV serotype 9 to deliver sFLT1 through intravenous injection (IV) to the brain angiogenic region. AAVs were packaged in AAV serotypes 1 and 2 (stereotactic injection) and 9 (IV injection). Brain angiogenesis was induced in adult mice through stereotactic injection of AAV1-VEGF. AAV2-sFLT02 containing sFLT1 VEGF-binding domain (domain 2) was injected into the brain angiogenic region, and AAV9-sFLT1 was injected into the jugular vein at the time of or 4 weeks after AAV1-VEGF injection. We showed that AAV2-sFLT02 inhibited brain angiogenesis at both time points. IV injection of AAV9-sFLT1 inhibited angiogenesis only when the vector was injected 4 weeks after angiogenic induction. Neither lymphocyte infiltration nor neuron loss was observed in AAV9-sFLT1-treated mice. Our data show that systemically delivered AAV9-sFLT1 inhibits angiogenesis in the mouse brain, which could be utilized to treat brain angiogenic diseases such as brain arteriovenous malformation.


Assuntos
Inibidores da Angiogênese/genética , Encéfalo/irrigação sanguínea , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Terapia Genética , Vetores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/terapia , Distribuição Aleatória , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/biossíntese
2.
Gene Ther ; 16(1): 10-6, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18633446

RESUMO

Vascular endothelial growth factor (VEGF) is important in pathological neovascularization, which is a key component of diseases such as the wet form of age-related macular degeneration, proliferative diabetic retinopathy and cancer. One of the most potent naturally occurring VEGF binders is VEGF receptor Flt-1. We have generated two novel chimeric VEGF-binding molecules, sFLT01 and sFLT02, which consist of the second immunoglobulin (IgG)-like domain of Flt-1 fused either to a human IgG1 Fc or solely to the CH3 domain of IgG1 Fc through a polyglycine linker 9Gly. In vitro analysis showed that these novel molecules are high-affinity VEGF binders. We have demonstrated that adeno-associated virus serotype 2 (AAV2)-mediated intravitreal gene delivery of sFLT01 efficiently inhibits angiogenesis in the mouse oxygen-induced retinopathy model. There were no histological observations of toxicity upon persistent ocular expression of sFLT01 for up to 12 months following intravitreal AAV2-based delivery in the rodent eye. Our data suggest that AAV2-mediated intravitreal gene delivery of our novel molecules may be a safe and effective treatment for retinal neovascularization.


Assuntos
Terapia Genética/métodos , Proteínas Recombinantes de Fusão/genética , Retina/metabolismo , Neovascularização Retiniana/terapia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Dependovirus/genética , Imunofluorescência , Expressão Gênica , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/análise , Proteínas Recombinantes de Fusão/metabolismo , Neovascularização Retiniana/metabolismo , Transdução Genética/métodos , Transgenes , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
3.
Cancer Res ; 61(7): 2983-95, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306477

RESUMO

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.


Assuntos
Arabinofuranosiluracila/análogos & derivados , Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/fisiologia , Transgenes , Animais , Arabinofuranosiluracila/farmacocinética , Autorradiografia , Chlorocebus aethiops , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glioma/genética , Gliossarcoma/genética , Herpesvirus Humano 1/genética , Humanos , Radioisótopos do Iodo , Óperon Lac/genética , Camundongos , Camundongos Nus , Mutação , Ratos , Timidina Quinase/biossíntese , Timidina Quinase/genética , Tomografia Computadorizada de Emissão , Células Vero , Replicação Viral
4.
J Gene Med ; 2(5): 379-89, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11045432

RESUMO

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.


Assuntos
Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Herpesvirus Humano 1/genética , Tetraciclina/farmacologia , Animais , Chlorocebus aethiops , Genes Precoces , Proteínas Imediatamente Precoces/genética , Luciferases/genética , Mutação , Plasmídeos/genética , Regiões Promotoras Genéticas , Transcrição Gênica , Transfecção , Células Vero , Proteínas Virais/metabolismo
5.
Eur J Biochem ; 267(8): 2195-207, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10759842

RESUMO

Stress can have profound effects on the cell. The elicitation of the stress response in the cell is often accompanied by the synthesis of high-molecular-mass complexes, sometimes termed heat shock granules (HSGs). The presence of the complexes has been shown to be important for the survival of cells subjected to stress. We purified these complexes from heat-stressed BY-2 tobacco cells. HSG complexes formed in vivo contain predominantly smHSPs, HSP40 and HSP70 and display chaperone-like activity. Tubulins as well as other proteins may be part of the complex or its substrate. The proteins, except smHSPs and to some extent HSP70, were hypersensitive to proteolysis, suggesting that they were partially denatured and not an integral part of the HSG complexes. When citrate synthase was used as the substrate, in vivo generated HSG complexes exhibited strong nucleotide-dependent in vitro chaperone activity. Measurable ATP-mediated hydrolytic activity was detected. Isolated HSG complexes are stable until ATP is added, which leads to rapid dissociation of the complex into subunits. It is proposed that smHSPs form the core of the complex in association with ATP-dependent HSP70 and HSP40 cochaperones. Implications of these findings are discussed.


Assuntos
Proteínas de Choque Térmico HSP70/análise , Proteínas de Choque Térmico/análise , Chaperonas Moleculares/química , Proteínas de Plantas/química , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Citrato (si)-Sintase/metabolismo , Eletroforese em Gel de Poliacrilamida , Endopeptidase K/metabolismo , Ativação Enzimática , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico/química , Immunoblotting , Cinética , Substâncias Macromoleculares , Microscopia Eletrônica , Plantas Tóxicas , Espalhamento de Radiação , Temperatura , Nicotiana
6.
J Virol ; 73(7): 6048-55, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364357

RESUMO

Toxicity and immunity associated with adenovirus backbone gene expression is an important hurdle to overcome for successful gene therapy. Recent efforts to improve adenovirus vectors for in vivo use have focused on the sequential deletion of essential early genes. Adenovirus vectors have been constructed with the E1 gene deleted and with this deletion in combination with an E2a, E2b, or E4 deletion. We report here a novel vector (Av4orf3nBg) lacking E1, E2a, and all of E4 except open reading frame 3 (ORF3) and expressing a beta-galactosidase reporter gene. This vector was generated by transfection of a plasmid carrying the full-length vector sequence into A30.S8 cells that express E1 and E2a but not E4. Production was subsequently performed in an E1-, E2a-, and E4-complementing cell line. We demonstrated with C57BL/6 mice that the Av4orf3nBg vector effected gene transfer with an efficiency comparable to that of the Av3nBg (wild-type E4) vector but that the former exhibited a higher level of beta-galactosidase expression. This observation suggests that E4 ORF3 alone is able to enhance RNA levels from the beta-galactosidase gene when the Rous sarcoma virus promoter is used to drive transgene expression in the mouse liver. In addition, we observed less liver toxicity in mice injected with the Av4orf3nBg vector than those injected with the Av3nBg vector at a comparable DNA copy number per cell. This study suggests that the additional deletion of E4 in an E1 and E2a deletion background may be beneficial in decreasing immunogenicity and improving safety and toxicity profiles, as well as increasing transgene capacity and expression for liver-directed gene therapy.


Assuntos
Proteínas E1 de Adenovirus/genética , Proteínas E2 de Adenovirus/genética , Proteínas E3 de Adenovirus/genética , Proteínas E4 de Adenovirus/genética , Adenovírus Humanos/genética , Vetores Genéticos/genética , Animais , Clonagem Molecular , Feminino , Deleção de Genes , Expressão Gênica , Humanos , Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese , Plasmídeos , Recombinação Genética , Transgenes
7.
Hum Gene Ther ; 10(2): 311-8, 1999 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-10022555

RESUMO

This study investigated the intraarterial delivery of genetically engineered replication-deficient adenovirus vectors (AVs) and cationic liposome-plasmid DNA complexes (lipoDNA) to experimental brain tumors. Adenovirus or lipoDNA was injected into the internal carotid artery (ICA) of F344 rats harboring intracerebral 9L gliosarcomas, using bradykinin (BK) to selectively permeabilize the blood-tumor barrier (BTB). Brain and internal organs of the animals were collected 48 hr after vector injection and stained for expression of the marker gene product, beta-galactosidase (beta-Gal). Intracarotid delivery of AV to 9L rat gliosarcoma without BTB disruption resulted in transgene expression in 3-10% of tumor cells distributed throughout the tumor. Virus-mediated expression of beta-gal gene products in this tumor model was particularly high in small foci (< or = 0.5 mm), which had invaded the normal brain tissue surrounding the main tumor mass. In these foci more than 50% of tumor cells were transduced. BK infusion increased the amount of transgene-expressing cells in larger tumor foci to 15-30%. In the brain parenchyma only a few endothelial cells expressed beta-gal owing to AV-mediated gene transfer. Intracarotid delivery of lipoDNA bearing a cytoplasmic expression cassette rendered more than 30% of the tumor cells positive for the marker gene without BTB disruption. The pattern of distribution was in general homogeneous throughout the tumor. BK infusion was able to increase further the number of transduced tumor cells to more than 50%. Although lipoDNA-mediated gene transfer showed increased efficacy as compared with AV-mediated gene transfer, it had less specificity since a larger number of endothelial and glial cells also expressed the transgene. AV and lipoDNA injections, in the absence and presence of BK, also resulted in transduction of peripheral organs. AV showed its known predilection for liver and lung. In the case of lipoDNA, parenchymal organs such as liver, lung, testes, lymphatic nodes, and especially spleen, were transduced. These findings indicate that intracarotid application of AV and lipoDNA vectors can effectively transduce tumor cells in the brain, and that BTB modulation by BK infusion can further increase the number of transgene-expressing tumor cells.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/terapia , DNA/administração & dosagem , Vetores Genéticos , Gliossarcoma/terapia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Bradicinina/farmacologia , Neoplasias Encefálicas/metabolismo , Endotélio Vascular/metabolismo , Terapia Genética , Gliossarcoma/metabolismo , Injeções Intra-Arteriais , Masculino , Ratos , Ratos Endogâmicos F344
8.
J Gene Med ; 1(3): 176-85, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10738566

RESUMO

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.


Assuntos
Terapia Genética , Vetores Genéticos , Gliossarcoma/terapia , Herpesvirus Humano 1/genética , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Genes Precoces , Vírus Auxiliares/genética , Vírus Auxiliares/fisiologia , Herpesvirus Humano 1/fisiologia , Óperon Lac , Masculino , Camundongos , Camundongos Nus , Plasmídeos/genética , Ratos , Recombinação Genética , Células Vero , Replicação Viral
9.
Gene Ther ; 5(6): 809-19, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9747461

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Glioma/terapia , Simplexvirus/imunologia , Animais , Neoplasias Encefálicas/imunologia , Expressão Gênica , Marcadores Genéticos , Glioma/imunologia , Imunização , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo
10.
J Neurovirol ; 4(2): 133-47, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9584951

RESUMO

Gene therapy offers significant advantages to the field of oncology with the addition of specifically and uniquely engineered mechanisms of halting malignant proliferation through cytotoxicity or reproductive arrest. To confer a true benefit to the therapeutic ratio (the relative toxicity to tumor compared to normal tissue) a vector or the transgene it carries must selectively affect or access tumor cells. Beyond the selective toxicities of many transgene products, which frequently parallel that of contemporary chemotherapeutic agents, lies the potential utility of targeting the vector. This review presents an overview of current and potential methods for designing vectors targeted to CNS malignancies through selective delivery, cell entry, transport or transcriptional regulation. The topic of delivery encompasses physical and pharmaceutic means of increasing the relative exposure of tumors to vector. Cell entry based methodologies are founded on increasing relative uptake of vector through the chemical or recombinant addition of ligand and antibody domains which selectively bind receptors expressed on target cells. Targeted transport involves the potential for using cells to selectively carry vectors or transgenes into tumors. Finally, promoter and enhancer systems are discussed which have potential for selectivity activating transcription to produce targeted transgene expression or vector propagation.


Assuntos
Neoplasias do Sistema Nervoso Central/terapia , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Glioma/terapia , Humanos , Transcrição Gênica , Replicação Viral
11.
Hum Gene Ther ; 8(17): 2057-68, 1997 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-9414254

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética , Vetores Genéticos , Gliossarcoma/terapia , Herpesvirus Humano 1/genética , Ribonucleotídeo Redutases/genética , Proteínas Virais/genética , Animais , Chlorocebus aethiops , Terapia Combinada , Modelos Animais de Doenças , Ganciclovir/uso terapêutico , Deleção de Genes , Vetores Genéticos/toxicidade , Gliossarcoma/secundário , Humanos , Masculino , Neoplasias Meníngeas/secundário , Ratos , Ratos Endogâmicos F344 , Ribonucleotídeo Redutases/metabolismo , Células Tumorais Cultivadas , Células Vero , Proteínas Virais/metabolismo , Replicação Viral
12.
Hum Gene Ther ; 8(3): 331-9, 1997 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-9048200

RESUMO

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.


Assuntos
Astrócitos/metabolismo , Astrócitos/transplante , Transplante de Células/métodos , Córtex Cerebral/citologia , Terapia Genética/métodos , Fatores de Crescimento Neural/biossíntese , Linhagem Celular , Células Cromafins , Feto , Vetores Genéticos/genética , Humanos , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Retroviridae , Transfecção/genética , Transfecção/métodos
13.
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
14.
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
15.
Hum Gene Ther ; 7(16): 1989-94, 1996 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-8930659

RESUMO

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.


Assuntos
Antivirais/farmacologia , Neoplasias Encefálicas/terapia , Ganciclovir/farmacologia , Vetores Genéticos/fisiologia , Gliossarcoma/terapia , Herpesvirus Humano 1/fisiologia , Animais , Modelos Animais de Doenças , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Humanos , Injeções Espinhais , Neoplasias Experimentais , Ratos , Timidina Quinase/genética , Fatores de Tempo , Células Tumorais Cultivadas
16.
Hum Gene Ther ; 7(16): 2003-13, 1996 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-8930661

RESUMO

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.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Vírus Auxiliares/genética , Herpesvirus Humano 1/genética , Animais , Southern Blotting , Linhagem Celular , Chlorocebus aethiops , Humanos , Plasmídeos , Células Vero
17.
Hum Gene Ther ; 7(3): 291-300, 1996 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-8835217

RESUMO

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.


Assuntos
Neoplasias do Sistema Nervoso Central/terapia , Cisterna Magna , Lobo Frontal , Genes Reporter , Vetores Genéticos/genética , Gliossarcoma/secundário , Neoplasias Meníngeas/secundário , Simplexvirus/genética , Transfecção , Animais , Neoplasias do Sistema Nervoso Central/patologia , Genes Sintéticos , Gliossarcoma/patologia , Gliossarcoma/terapia , Injeções Espinhais , Masculino , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/terapia , Metástase Neoplásica , Transplante de Neoplasias , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/análise , Simplexvirus/patogenicidade , Simplexvirus/fisiologia , Espaço Subaracnóideo , Timidina Quinase/análise , Timidina Quinase/biossíntese , Timidina Quinase/genética , Replicação Viral , beta-Galactosidase/análise , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
18.
Brain Pathol ; 5(4): 345-81, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8974620

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética , Formação de Anticorpos , Ensaios Clínicos como Assunto , Terapia Genética/métodos , Vetores Genéticos , Humanos , Neovascularização Patológica , Transgenes , Vírus/genética
19.
Neuroreport ; 6(4): 669-72, 1995 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-7605924

RESUMO

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.


Assuntos
Fibroblastos/metabolismo , Terapia Genética , Ataque Isquêmico Transitório/tratamento farmacológico , Fatores de Crescimento Neural/farmacologia , Prosencéfalo/irrigação sanguínea , Animais , Linhagem Celular , Fibroblastos/transplante , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar
20.
Neurosci Lett ; 153(1): 111-4, 1993 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-8099723

RESUMO

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.


Assuntos
Astrócitos/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Glutamatos/farmacologia , Fatores de Crescimento Neural/genética , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Animais , Northern Blotting , Células Cultivadas , Expressão Gênica , Ácido Glutâmico , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...