Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Gene Ther ; 22(2): 181-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25474439

RESUMO

Sandhoff disease (SD) is caused by deficiency of N-acetyl-ß-hexosaminidase (Hex) resulting in pathological accumulation of GM2 ganglioside in lysosomes of the central nervous system (CNS) and progressive neurodegeneration. Currently, there is no treatment for SD, which often results in death by the age of five years. Adeno-associated virus (AAV) gene therapy achieved global CNS Hex restoration and widespread normalization of storage in the SD mouse model. Using a similar treatment approach, we sought to translate the outcome in mice to the feline SD model as an important step toward human clinical trials. Sixteen weeks after four intracranial injections of AAVrh8 vectors, Hex activity was restored to above normal levels throughout the entire CNS and in cerebrospinal fluid, despite a humoral immune response to the vector. In accordance with significant normalization of a secondary lysosomal biomarker, ganglioside storage was substantially improved, but not completely cleared. At the study endpoint, 5-month-old AAV-treated SD cats had preserved neurological function and gait compared with untreated animals (humane endpoint, 4.4±0.6 months) demonstrating clinical benefit from AAV treatment. Translation of widespread biochemical disease correction from the mouse to the feline SD model provides optimism for treatment of the larger human CNS with minimal modification of approach.


Assuntos
Terapia Genética , Doença de Sandhoff/terapia , Animais , Gatos , Dependovirus/genética , Dependovirus/imunologia , Progressão da Doença , Gangliosídeos/metabolismo , Vetores Genéticos , Humanos , Imunidade Humoral , Injeções Intraventriculares , Doença de Sandhoff/patologia , Transdução Genética , Resultado do Tratamento , beta-N-Acetil-Hexosaminidases/biossíntese , beta-N-Acetil-Hexosaminidases/genética
2.
Gene Ther ; 21(12): 1041-50, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25273354

RESUMO

Familial amyloidotic polyneuropathy (FAP) is a neurodegenerative disorder characterized by extracellular deposition of amyloid fibrils composed by mutated transthyretin (TTR) mainly in the peripheral nervous system. At present, liver transplantation is still the standard treatment to halt the progression of clinical symptoms in FAP, but new therapeutic strategies are emerging, including the use of TTR stabilizers. Here we propose to establish a new gene therapy approach using adeno-associated virus (AAV) vectors to deliver the trans-suppressor TTR T119M variant to the liver of transgenic TTR V30M mice at different ages. This TTR variant is known for its ability to stabilize the tetrameric protein. Analysis of the gastrointestinal tract of AAV-treated animals revealed a significant reduction in deposition of TTR non-fibrillar aggregates in as much as 34% in stomach and 30% in colon, as well as decreased levels of biomarkers associated with TTR deposition, namely the endoplasmic reticulum stress marker BiP and the extracellular matrix protein MMP-9. Moreover, we showed with different studies that our approach leads to an increase in tetrameric and more stable forms of TTR, in favor of destabilized monomers. Altogether our data suggest the possibility to use this gene therapy approach in a prophylactic manner to prevent FAP pathology.


Assuntos
Neuropatias Amiloides Familiares/terapia , Terapia Genética/métodos , Pré-Albumina/genética , Neuropatias Amiloides Familiares/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Estresse do Retículo Endoplasmático/genética , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Marcadores Genéticos , Vetores Genéticos , Fígado/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Sistema Nervoso Periférico/metabolismo , Pré-Albumina/metabolismo , Proteômica
3.
Gene Ther ; 18(12): 1173-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21900963

RESUMO

Adeno-associated virus (AAV)-mediated gene replacement for lysosomal disorders have been spurred by the ability of some serotypes to efficiently transduce neurons in the brain and by the ability of lysosomal enzymes to cross-correct among cells. Here, we explored enzyme replacement therapy in a knock-out mouse model of congenital neuronal ceroid lipofuscinosis (NCL), the most severe of the NCLs in humans. The missing protease in this disorder, cathepsin D (CathD) has high levels in the central nervous system. This enzyme has the potential advantage for assessing experimental therapy in that it can be imaged using a near-infrared fluorescence (NIRF) probe activated by CathD. Injections of an AAV2/rh8 vector-encoding mouse CathD (mCathD) into both cerebral ventricles and peritoneum of newborn knock-out mice resulted in a significant increase in lifespan. Successful delivery of active CathD by the AAV2/rh8-mCathD vector was verified by NIRF imaging of mouse embryonic fibroblasts from knock-out mice in culture, as well as by ex vivo NIRF imaging of the brain and liver after gene transfer. These studies support the potential effectiveness and imaging evaluation of enzyme replacement therapy to the brain and other organs in CathD null mice via AAV-mediated gene delivery in neonatal animals.


Assuntos
Catepsina D/genética , Corantes Fluorescentes , Técnicas de Transferência de Genes , Terapia Genética/métodos , Raios Infravermelhos , Lipofuscinoses Ceroides Neuronais/terapia , Animais , Animais Recém-Nascidos , Química Encefálica , Dependovirus/genética , Modelos Animais de Doenças , Terapia de Reposição de Enzimas/métodos , Vetores Genéticos , Fígado/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Lipofuscinoses Ceroides Neuronais/genética
4.
Gene Ther ; 16(2): 303-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18818671

RESUMO

GM1-gangliosidosis is a lysosomal storage disease (LSD) caused by an autosomal recessive deficiency of lysosomal acid beta-galactosidase (betagal). This leads to accumulation of GM1-ganglioside and its asialo derivative GA1 in the central nervous system (CNS), and progressive neurodegeneration. Therapeutic AAV-mediated gene delivery to the brain for LSDs has proven very successful in several animal models. GM1-gangliosidosis is also a prime candidate for AAV-mediated gene therapy in the CNS. As global neuropathology characterizes the most severe forms of this disease, therapeutic interventions need to achieve distribution of betagal throughout the entire CNS. Therefore, careful consideration of routes of administration and target structures from where metabolically active enzyme can be produced, released and distributed throughout the CNS, is necessary. The goal of this study was to investigate the pattern and mechanism of distribution of betagal in the adult GM1-gangliosidosis mouse brain upon hippocampal injection of an AAV vector-encoding betagal. We found evidence that three different mechanisms contribute to its distribution in the brain: (1) diffusion; (2) axonal transport within neurons from the site of production; (3) CSF flow in the perivascular space of Virchow-Robin. In addition, we found evidence of axonal transport of vector-encoded mRNA.


Assuntos
Encéfalo/enzimologia , Gangliosidose GM1/enzimologia , Terapia Genética/métodos , beta-Galactosidase/genética , Animais , Transporte Axonal , Dependovirus/genética , Modelos Animais de Doenças , Gangliosidose GM1/terapia , Vetores Genéticos/farmacocinética , Hipocampo/enzimologia , Camundongos , Camundongos Knockout , Neurônios/fisiologia , RNA Mensageiro/genética , Distribuição Tecidual , beta-Galactosidase/biossíntese , beta-Galactosidase/deficiência , beta-Galactosidase/farmacocinética
5.
Mol Ther ; 15(1): 30-7, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17164772

RESUMO

GM1-gangliosidosis is a glycosphingolipid (GSL) lysosomal storage disease caused by autosomal recessive deficiency of lysosomal acid beta-galactosidase (betagal), and characterized by accumulation of GM1-ganglioside and GA1 in the brain. Here we examined the effect of neonatal intracerebroventricular (i.c.v.) injection of an adeno-associated virus (AAV) vector encoding mouse betagal on enzyme activity and brain GSL content in GM1-gangliosidosis (betagal(-/-)) mice. Histological analysis of betagal distribution in 3-month-old AAV-treated betagal(-/-) mice showed that enzyme was present at high levels throughout the brain. Biochemical quantification showed that betagal activity in AAV-treated brains was 7- to 65-fold higher than in wild-type controls and that brain GSL levels were normalized. Cerebrosides and sulfatides, which were reduced in untreated betagal(-/-) mice, were restored to normal levels by AAV treatment. In untreated betagal(-/-) brains, cholesterol was present at normal levels but showed abnormal cellular distribution consistent with endosomal/lysosomal localization. This feature was also corrected in AAV-treated mice. The biochemical and histological parameters analyzed in this study showed that normal brain neurochemistry was achieved in AAV-treated betagal(-/-) mice. Therefore we show for the first time that neonatal AAV-mediated gene delivery of lysosomal betagal to the brain may be an effective approach for treatment of GM1-gangliosidosis.


Assuntos
Dependovirus/genética , Gangliosidose GM1/genética , Gangliosidose GM1/terapia , Terapia Genética , Lisossomos/enzimologia , beta-Galactosidase/deficiência , beta-Galactosidase/metabolismo , Animais , Animais Recém-Nascidos , Cromatografia Líquida de Alta Pressão , Gangliosidose GM1/enzimologia , Gangliosidose GM1/patologia , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , beta-Galactosidase/genética
6.
Neuroscience ; 138(2): 501-10, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16414198

RESUMO

Adeno-associated virus (AAV) vectors have gained a preeminent position in the field of gene delivery to the normal brain through their ability to achieve extensive transduction of neurons and to mediate long-term gene expression with no apparent toxicity. In adult animals direct infusion of AAV vectors into the brain parenchyma results in highly efficient transduction of target structures. However AAV-mediated global delivery to the adult brain has been an elusive goal. In contrast, widespread global gene delivery has been obtained by i.c.v. injection of AAV1 or AAV2 in neonates. Among the novel AAV serotypes cloned and engineered for production of recombinant vectors, AAV8 has shown a tremendous potential for in vivo gene delivery with nearly complete transduction of many tissues in rodents after intravascular infusion. Here we compare the efficiency of an AAV8 serotyped vector with that of AAV1 and AAV2 serotyped vectors for the extent of gene delivery to the brain after neonatal injection into the lateral ventricles. The vectors all encoded green fluorescent protein (GFP) under control of a hybrid CMV enhancer/chicken beta-actin promoter with AAV2 inverted terminal repeats, but differed from each other with respect to the capsid type. A total of 6.8 x 10(10) genome copies were injected into the lateral ventricles of postnatal day 0 mice. Mice were killed at postnatal day 30 and brains analyzed for distribution of GFP-positive cells. AAV8 proved to be more efficient than AAV1 or AAV2 vectors for gene delivery to all of the structures analyzed, including the cerebral cortex, hippocampus, olfactory bulb, and cerebellum. Moreover the intensity of gene expression, assessed using a microarray reader, was considerably higher for AAV8 in all structures analyzed. In conclusion, the enhanced transduction achieved by AAV8 compared with AAV1 and AAV2 indicates that AAV8 is the superior serotype for gene delivery to the CNS.


Assuntos
Encéfalo/fisiologia , Dependovirus/genética , Transfecção/métodos , Animais , Animais Recém-Nascidos , Sequência de Bases , Encéfalo/crescimento & desenvolvimento , Capsídeo , Linhagem Celular , Cerebelo/fisiologia , Córtex Cerebral/fisiologia , Primers do DNA , Dependovirus/classificação , Vetores Genéticos , Hipocampo/fisiologia , Humanos , Rim , Camundongos , Camundongos Endogâmicos C57BL , Bulbo Olfatório , Sorotipagem
7.
Cancer Res ; 56(23): 5423-7, 1996 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8968096

RESUMO

Interleukin 1beta-converting enzyme (ICE) is a member of a growing family of cysteine proteases shown to be a crucial component in the activation of a genetic program that leads to autonomous cell death in mammalian cells. In this study, a murine ICE-lacZ fusion gene was introduced into a novel retroviral vector designed to achieve regulated ectopic expression of a foreign gene in mammalian cells. By delivering the ICE-lacZ gene within a retroviral vector and under the control of a tetracycline-regulated promoter, we were able to utilize the intrinsic cell death program of ICE as a means for tumoricidal therapy in a rat brain tumor model. Both in culture and in vivo suppression of ICE-lacZ expression was extremely tight in the presence of tetracycline, as determined by the lack of X-galactosidase-positive tumor cells and by cell viability. When tetracycline was withdrawn, ICE-lacZ gene expression was rapidly turned on and apoptosis-mediated cell death occurred in essentially all tumor cells.


Assuntos
Apoptose/fisiologia , Neoplasias Encefálicas/terapia , Cisteína Endopeptidases/fisiologia , Terapia Genética , Vetores Genéticos/genética , Gliossarcoma/terapia , Vírus da Leucemia Murina de Moloney/genética , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Caspase 1 , Corpo Estriado , Cisteína Endopeptidases/biossíntese , Cisteína Endopeptidases/genética , Indução Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Gliossarcoma/patologia , Masculino , Transplante de Neoplasias , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Tetraciclina/farmacologia , Tetraciclina/uso terapêutico , Transfecção
8.
Cancer Res ; 54(22): 5745-51, 1994 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-7954393

RESUMO

Survival of rats harboring cerebral 9L gliosarcomas can be significantly extended by an intratumoral inoculation with a herpes simplex virus vector, designated as hrR3. This vector, which bears the lacZ reporter gene, is defective in the gene encoding ribonucleotide reductase, allowing for replication in dividing tumor cells but not in postmitotic neural cells. It also possesses an intact viral thymidine kinase (TK) gene, which confers chemosensitivity to ganciclovir. In this study, the ability of ganciclovir to potentiate the antitumor effect of hrR3 was evaluated. In culture, there was a 23% decrease in the growth of 9L cells treated with hrR3 plus ganciclovir compared to hrR3 alone (P < 0.01). The combination of hrR3 plus ganciclovir led to the long-term survival of 48% of rats harboring intracerebral 9L gliosarcomas compared to 20% survival in the hrR3 group (P < 0.05). Ganciclovir treatment had no effect on the growth of tumor cells in vitro or in vivo when a herpes simplex virus vector with a defective TK gene was used. Immunocytochemistry confirmed selective expression of the TK gene in cells within the tumor. These findings indicate that the TK gene can potentiate the antitumor effect of the hrR3 herpes simplex virus vector and provide the basis for placing additional therapeutic genes in the genome of hrR3.


Assuntos
Neoplasias Encefálicas/terapia , Ganciclovir/uso terapêutico , Terapia Genética/métodos , Gliossarcoma/terapia , Simplexvirus/genética , Timidina Quinase/genética , Animais , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Terapia Combinada , Vetores Genéticos/genética , Gliossarcoma/enzimologia , Gliossarcoma/genética , Gliossarcoma/mortalidade , Gliossarcoma/patologia , Masculino , Ratos , Ratos Endogâmicos F344 , Simplexvirus/enzimologia , Timidina Quinase/análise , Células Tumorais Cultivadas
9.
Hum Gene Ther ; 12(14): 1771-83, 2001 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-11560770

RESUMO

Mutations in the alpha-chain of lysosomal hexosaminidase (EC 3.2.1.52) underlie two distinct biochemical phenotypes known as variant B and variant B1 of G(M2) gangliosidosis. This paper shows that the transduction of human B1-type fibroblasts (producing catalytically inactive alpha-chains) with a retroviral vector encoding the human hexosaminidase alpha-chain leads to a complete correction of HexA (alpha beta dimer) activity with both synthetic and natural substrates. The alpha-subunit overexpression leads to a partial HexB (beta beta dimer) depletion corresponding to about 10% of control HexB activity. The newly synthesized enzyme is correctly processed and targeted to the lysosomes in transduced cells. The high levels of recombinant enzyme correctly produced the metabolic defect, enabling the cells efficiently to degrade the accumulated storage product present in lysosomes. The transduced fibroblasts are also able to secrete HexA efficiently into the culture medium. Moreover, transfer of the human transgene product to B1-type deficient fibroblasts lead to an increase of activity against 4MUGS, the alpha-chain specific synthetic substrate, up to 30% of the control mean activity level. This level of activity might be sufficient to restore the normal ganglioside G(M2) metabolism in recipient cells. The data obtained demonstrate that B1-type phenotype can be efficiently corrected by retrovirus-mediated gene transfer.


Assuntos
DNA Complementar/metabolismo , Fibroblastos/metabolismo , Gangliosídeo G(M2)/genética , Gangliosidoses GM2/genética , Técnicas de Transferência de Genes , Retroviridae/genética , beta-N-Acetil-Hexosaminidases/genética , Células 3T3 , Animais , Linhagem Celular , Dimerização , Eletroforese em Gel de Poliacrilamida , Gangliosídeo G(M2)/metabolismo , Gangliosidoses GM2/metabolismo , Vetores Genéticos , Hexosaminidase A , Hexosaminidase B , Humanos , Imunoglobulina M/metabolismo , Lisossomos/metabolismo , Camundongos , Microscopia de Fluorescência , Mutação , Fenótipo , Testes de Precipitina , Proteínas Recombinantes/metabolismo , Temperatura , Fatores de Tempo , Transdução Genética , Transgenes , beta-N-Acetil-Hexosaminidases/química
10.
Hum Gene Ther ; 11(5): 715-27, 2000 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-10757351

RESUMO

Mutations in the lysosomal acid beta-galactosidase (EC 3.2.1.23) underlie two different disorders: GM1 gangliosidosis, which involves the nervous system and visceral organs to varying extents, and Morquio's syndrome type B (Morquio B disease), which is a skeletal-connective tissue disease without any CNS symptoms. This article shows that transduction of human GM1 gangliosidosis fibroblasts with retrovirus vectors encoding the human acid beta-galactosidase cDNA leads to complete correction of the enzymatic deficiency. The newly synthesized enzyme is correctly processed and targeted to the lysosomes in transduced cells. Cross-correction experiments using retrovirus-modified cells as enzyme donors showed, however, that the human enzyme is transferred at low efficiencies. Experiments using a different retrovirus vector carrying the human cDNA confirmed this observation. Transduction of human GM1 fibroblasts and mouse NIH 3T3 cells with a retrovirus vector encoding the mouse beta-galactosidase cDNA resulted in high levels of enzymatic activity. Furthermore, the mouse enzyme was found to be transferred to human cells at high efficiency. Enzyme activity measurements in medium conditioned by genetically modified cells suggest that the human beta-galactosidase enzyme is less efficiently released to the extracellular space than its mouse counterpart. This study suggests that lysosomal enzymes, contrary to the generalized perception in the field of gene therapy, may differ significantly in their properties and provides insights for design of future gene therapy interventions in acid beta-galactosidase deficiency.


Assuntos
Gangliosidose GM1/enzimologia , Técnicas de Transferência de Genes , Retroviridae/genética , beta-Galactosidase/deficiência , Células 3T3/enzimologia , Células 3T3/virologia , Animais , Meios de Cultivo Condicionados , Fibroblastos/enzimologia , Fibroblastos/virologia , Gangliosidose GM1/genética , Vetores Genéticos , Humanos , Lisossomos/metabolismo , Camundongos , beta-Galactosidase/genética
11.
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
12.
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
13.
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
14.
Hum Gene Ther ; 9(9): 1261-73, 1998 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-9650611

RESUMO

Vector-mediated transfer of prodrug-activating genes provides a promising means of cancer gene therapy. In a search for more selective and more potent bioactivating enzymes for gene therapy of malignant brain tumors, the toxicity-generating capacity of the rabbit cytochrome P450 isozyme CYP4B1 was investigated. Rabbit CYP4B1, but not rat or human isozymes, efficiently converts the inert prodrugs, 2-aminoanthracene (2-AA) and 4-ipomeanol (4-IM), into highly toxic alkylating metabolites. Toxicity of these two prodrugs was evaluated in culture in parental and genetically modified rodent (9L) and human (U87) glioma cell lines stably expressing CYP4B1, and in vivo in a subcutaneous 9L tumor model in nude mice. The most sensitive CYP4B1-expressing glioma clone, 9L4B1-60, displayed an LD50 of 2.5 microM for 2-AA and 4-IM after 48 h of prodrug incubation, whereas 20 times higher prodrug concentrations did not cause any significant toxicity to control cells. Substantial killing of control tumor cells by 2-AA was achieved by co-culturing these cells with CYP4B1-expressing cells at a ratio of 100:1, and toxic metabolites could be transferred through medium. In both CYP4B1-expressing cells and co-cultured control cells, prodrug bioactivation was associated with DNA fragmentation, as assayed by fluorescent TUNEL assays and by annexin V staining. Alkaline elution of cellular DNA after exposure to 4-IM revealed extensive protein-DNA crosslinking with single-strand breakage. Growth of 9L-4B1 tumors in nude mice was inhibited by intraperitoneal injection of 4-IM with minimal side effects. Potential advantages of the CYP4B1 gene therapy paradigm include: the low concentrations of prodrug needed to kill sensitized tumor cells; low prodrug conversion by human isozymes, thus reducing toxicity to normal cells; a tumor-killing bystander effect that can occur even without cell-to-cell contact; and the utilization of lipophilic prodrugs that can penetrate the blood-brain barrier.


Assuntos
Antracenos/farmacologia , Antineoplásicos/farmacologia , Hidrocarboneto de Aril Hidroxilases , Neoplasias Encefálicas/terapia , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , DNA de Neoplasias/efeitos dos fármacos , Terapia Genética/métodos , Glioblastoma/terapia , Gliossarcoma/terapia , Pró-Fármacos/farmacologia , Terpenos/farmacologia , Animais , Apoptose , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Meios de Cultivo Condicionados , Dano ao DNA , Fragmentação do DNA , Glioblastoma/metabolismo , Glioblastoma/patologia , Gliossarcoma/metabolismo , Gliossarcoma/patologia , Glutationa/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Plasmídeos/genética , Coelhos , Ratos , Transfecção , Células Tumorais Cultivadas
15.
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
16.
Neoplasia ; 1(2): 154-61, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10933050

RESUMO

Current gene therapy technology is limited by the paucity of methodology for determining the location and magnitude of therapeutic transgene expression in vivo. We describe and validate a paradigm for monitoring therapeutic transgene expression by noninvasive imaging of the herpes simplex virus type 1 thymidine kinase (HSV-1-tk) marker gene expression. To test proportional coexpression of therapeutic and marker genes, a model fusion gene comprising green fluorescent protein (gfp) and HSV-1-tk genes was generated (tkgfp gene) and assessed for the functional coexpression of the gene product, TKGFP fusion protein, in rat 9L gliosarcoma, RG2 glioma, and W256 carcinoma cells. Analysis of the TKGFP protein demonstrated that it can serve as a therapeutic gene by rendering tkgfp transduced cells sensitive to ganciclovir or as a screening marker useful for identifying transduced cells by fluorescence microscopy or fluorescence-activated cell sorting (FACS). TK and GFP activities in the TKGFP fusion protein were similar to corresponding wild-type proteins and accumulation of the HSV-1-tk-specific radiolabeled substrate, 2'-fluoro-2'-deoxy-1beta-D-arabinofuranosyl-5-iodo-uracil (FIAU), in stability transduced clones correlated with gfp-fluorescence intensity over a wide range of expression levels. The tkgfp fusion gene itself may be useful in developing novel cancer gene therapy approaches. Valuable information about the efficiency of gene transfer and expression could be obtained by non-invasive imaging of tkgfp expression with FIAU and clinical imaging devices (gamma camera, positron-emission tomography [PET], single photon emission computed tomography [SPECT]), and/or direct visualization of gfp expression in situ by fluorescence microscopy or endoscopy.


Assuntos
Herpesvirus Humano 1/enzimologia , Proteínas Luminescentes/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Timidina Quinase/metabolismo , Transgenes/genética , Animais , Antivirais/farmacologia , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/farmacologia , Western Blotting , Separação Celular , Clonagem Molecular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Ganciclovir/farmacologia , Terapia Genética/métodos , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Microscopia de Fluorescência , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes de Fusão/genética , Retroviridae/metabolismo , Timidina Quinase/genética , Transdução Genética , Células Tumorais Cultivadas
17.
J Neuropathol Exp Neurol ; 59(7): 607-13, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10901232

RESUMO

A large animal tumor model for anaplastic glioma has been recently developed using immunotolerant allogeneic Beagle dogs and an established canine glioma cell line, J3T. This model offers advantages in terms of tumor morphology and similarity to human anaplastic glioma. The present study was aimed at evaluating the biological characteristics of the J3T canine glioma cell line as related to experimental gene therapy studies. Furthermore, development and morphology of canine brain tumors in a xenogeneic immunodeficient SCID mouse model was investigated. It was demonstrated that cultured J3T cells can be efficiently infected by adenovirus (AV), herpes-simplex type I (HSV), or retrovirus (RV) vectors, as well as by non-virus vectors such as cationic liposome/DNA complexes. Thus, in terms of infectability and transfectability, J3T cells seem to be closer to human glioma than the 9L rodent gliosarcoma. Cytotoxicity of selection antibiotics such as G418, puromycin, and hygromycin on J3T cells essentially resemble cytotoxicity seen with other established glioma lines, for example, 9L, U87, or U343. RV-mediated HSV-TK/GCV gene therapy demonstrated comparable LD50 for TK-expressing and control (non-expressing) J3T and 9L cells treated with Ganciclovir. Further, it was proven that J3T cells are tumorigenic and may grow heterotopically and orthotopically in a xenogeneic immunodeficient host, the SCID mouse, although morphology and growth pattern of these xenogeneic tumors differ from the demonstrated invasive phenotype in the Beagle dog.


Assuntos
Neoplasias Encefálicas , Técnicas de Cultura de Células/métodos , Cinamatos , Glioblastoma , Neoplasias Experimentais , Células 3T3 , Adenoviridae/genética , Animais , Antibacterianos/farmacologia , Antivirais/farmacologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Chlorocebus aethiops , Cães , Resistência Microbiana a Medicamentos , Ganciclovir/farmacologia , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Gliossarcoma , Herpesvirus Humano 1/genética , Humanos , Higromicina B/análogos & derivados , Higromicina B/farmacologia , Rim/citologia , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Ratos , Timidina Quinase/genética , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/transplante , Células Vero
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.
Neuroscience ; 125(3): 651-61, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15099679

RESUMO

TorsinA is a novel protein identified in the search for mutations underlying the human neurologic movement disorder, early onset torsion dystonia. Relatively little is understood about the normal function of torsinA or the physiological effects of the codon deletion associated with most cases of disease. Overexpression of wild-type torsinA in cultured cells by DNA transfection results in a reticular distribution of immunoreactive protein that co-localizes with endoplasmic reticulum resident chaperones, while the dystonia-related mutant form accumulates within concentric membrane whorls and nuclear-associated membrane stacks. In this study we examined the biogenesis of mutant torsinA-positive membrane inclusions using tetracycline-regulated herpes simplex virus amplicon vectors. At low expression levels, mutant torsinA was localized predominantly around the nucleus, while at high levels it was also concentrated within cytosolic spheroid inclusions. In contrast, the distribution of wild-type torsinA did not vary, appearing diffuse and reticular at all expression levels. These observations are consistent with descriptions of inducible membrane synthesis in other systems in which cytosolic membrane whorls are derived from multilayered membrane stacks that first form around the nuclear envelope. These results also suggest that formation of mutant torsinA-positive inclusions occurs at high expression levels in culture, whereas the perinuclear accumulation of the mutant protein is present even at low expression levels that are more likely to resemble those of the endogenous protein. These nuclear-associated membrane structures enriched in mutant torsinA may therefore be of greater relevance to understanding how the dystonia-related mutation compromises cellular physiology.


Assuntos
Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Corpos de Inclusão/metabolismo , Membranas Intracelulares/metabolismo , Chaperonas Moleculares/metabolismo , Organelas/metabolismo , Animais , Biomarcadores , Proteínas de Transporte/genética , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/patologia , Citosol/metabolismo , Citosol/patologia , Distonia Muscular Deformante/genética , Distonia Muscular Deformante/metabolismo , Distonia Muscular Deformante/fisiopatologia , Genes Reporter/genética , Vetores Genéticos/genética , Herpes Simples/genética , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/patologia , Membranas Intracelulares/patologia , Chaperonas Moleculares/genética , Mutação/genética , Membrana Nuclear/metabolismo , Membrana Nuclear/patologia , Organelas/genética , Organelas/patologia , Tetraciclina/farmacologia , Transgenes/genética
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA