Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 19(2): 182-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22113317

RESUMO

Insertional mutagenesis following gene therapy with gammaretroviral vectors can cause the development of lymphoproliferation in children with X-linked severe combined immunodeficiency. In experimental studies, recombinant adeno-associated virus (rAAV) vectors have also been reported to increase susceptibility to carcinogenesis. The possibility of vector-induced transformation in quiescent ocular cells is probably significantly lower than in mitotically active cells, but given the increasing number of clinical applications of rAAV and lentiviral vectors for ocular disease, a specific assessment of their oncogenic potential in the eye is important. In this study, we investigated the effect of rAAV2/2 and integrating HIV-1 vectors upon the incidence of ocular neoplasia in p53 tumour-suppressor gene-knockout (p53(-/-)) mice, which are highly susceptible to intraocular malignant transformation. Subretinal injections of high titre rAAV2/2 or integrating HIV-1 vectors induced no tumours in p53(-/-) or p53(+/-) animals, nor significantly affected their natural longevity. We conclude that any insertional events arising from subretinal delivery of these vectors appear insufficient to cause intraocular malignancy, even in highly susceptible animals. These findings support the continued development of these vectors for ocular applications.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes/efeitos adversos , Vetores Genéticos/efeitos adversos , Lentivirus/genética , Proteína Supressora de Tumor p53/genética , Animais , Transformação Celular Neoplásica/genética , Eletrorretinografia , Neoplasias Oculares/genética , Técnicas de Inativação de Genes , Terapia Genética , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde , Camundongos , Retina , Proteína Supressora de Tumor p53/deficiência
2.
Gene Ther ; 19(2): 145-53, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22052240

RESUMO

Substantial advances in our understanding of lentivirus lifecycles and their various constituent proteins have permitted the bioengineering of lentiviral vectors now considered safe enough for clinical trials for both lethal and non-lethal diseases. They possess distinct properties that make them particularly suitable for gene delivery in ophthalmic diseases, including high expression, consistent targeting of various post-mitotic ocular cells in vivo and a paucity of associated intraocular inflammation, all contributing to their ability to mediate efficient and stable intraocular gene transfer. In this review, the intraocular tropisms and therapeutic applications of both primate and non-primate lentiviral vectors, and how the unique features of the eye influence these, are discussed. The feasibility of therapeutic targeting using these vectors in animal models of both anterior and posterior ophthalmic disorders has been established, and has, in combination with substantial progress in enhancing lentiviral vector bio-safety over the past two decades, paved the way for the first human ophthalmic clinical trials using lentivirus-based gene transfer vectors.


Assuntos
Neovascularização de Coroide/genética , Técnicas de Transferência de Genes , Glaucoma/genética , Lentivirus , Degeneração Retiniana/genética , Animais , Neovascularização de Coroide/terapia , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Glaucoma/terapia , Humanos , Degeneração Retiniana/terapia
3.
Gene Ther ; 13(15): 1153-65, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16572190

RESUMO

We evaluated the efficacy of equine infectious anaemia virus (EIAV)-based lentiviral vectors encoding endostatin (EIAV.endostatin) or angiostatin (EIAV.angiostatin) in inhibiting angiogenesis and vascular hyperpermeability in the laser-induced model of choroidal neovascularisation (CNV). Equine infectious anaemia virus.endostatin, EIAV.angiostatin or control (EIAV.null) vectors were administered into the subretinal space of C57Bl/6J mice. Two weeks after laser injury CNV areas and the degree of vascular hyperpermeability were measured by image analysis of in vivo fluorescein angiograms. Compared with EIAV.null-injected eyes, EIAV.endostatin resulted in a 59.5% (P<0.001) reduction in CNV area and a reduction in hyperpermeability of 25.6% (P<0.05). Equine infectious anaemia virus.angiostatin resulted in a 50.0% (P<0.05) reduction in CNV area and a 23.9% (P<0.05) reduction in hyperpermeability. Equine infectious anaemia virus.endostatin, but not EIAV.angiostatin significantly augmented the frequency of apoptosis within the induced CNV as compared with injected controls. TdT-dUTP terminal nick end labeling analysis 5 weeks post-injection, and histological and retinal flatmount analysis 12 months post-injection revealed no evidence of vector- or transgene expression-related deleterious effects on neurosensory retinal cells, or mature retinal vasculature in non-lasered eyes. Highly expressing EIAV-based vectors encoding endostatin or angiostatin effectively control angiogenesis and hyperpermeability in experimental CNV without long-term deleterious effects, supporting the use of such a strategy in the management of patients with exudative age-related macular degeneration.


Assuntos
Angiostatinas/genética , Neovascularização de Coroide/terapia , Endostatinas/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vírus da Anemia Infecciosa Equina/genética , Inibidores da Angiogênese/genética , Animais , Apoptose , Permeabilidade Capilar , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/fisiopatologia , Angiofluoresceinografia , Vetores Genéticos/genética , Marcação In Situ das Extremidades Cortadas , Lasers , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neovascularização Patológica , Transdução Genética/métodos , Regulação para Cima
4.
J Gene Med ; 8(3): 275-85, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16299834

RESUMO

BACKGROUND: We have developed minimal non-primate lentiviral vectors based on the equine infectious anaemia virus (EIAV). We evaluated the in vivo expression profiles of these vectors delivered regionally to ocular tissues to define their potential utility in ocular gene therapy. METHODS: EIAV vectors pseudotyped with VSV-G or rabies-G envelope proteins were delivered subretinally, intravitreally or into the anterior chambers (intracameral administration) in mice. Reporter gene (eGFP) expression was analysed using in vivo retinal imaging or histological examination of eyes and brains at intervals between 3 days and 16 months. We investigated the effects of vector titre, pseudotype, genome configuration, site of intraocular administration, intentional retinal trauma and the degree of retinal maturation on the spatial and temporal expression profiles of these vectors. RESULTS: Subretinal vector delivery resulted in efficient and stable transduction of retinal pigment epithelial (RPE) cells and variable transduction of photoreceptors up to 16 months post-injection. Retinal trauma facilitated the local transduction of neurosensory retinal cells. Intracameral administration of VSV-G- but not rabies-G-pseudotyped vectors produced stable eGFP expression in corneal endothelial cells and trabecular meshwork. CONCLUSIONS: The cellular tropism and expression kinetics of optimised EIAV vectors after intraocular administration make them attractive vehicles for delivering therapeutic genes in the management of inherited and acquired retinal and anterior segment disorders.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Vírus da Anemia Infecciosa Equina/genética , Doenças Retinianas/terapia , Animais , Feminino , Perfilação da Expressão Gênica , Genes Reporter , Vetores Genéticos , Genoma , Proteínas de Fluorescência Verde/biossíntese , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Células Fotorreceptoras , Epitélio Pigmentado Ocular/fisiologia , Retina/lesões , Doenças Retinianas/genética , Transdução Genética , Tropismo
5.
Gene Ther ; 12(8): 694-701, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15660111

RESUMO

The Royal College of Surgeons (RCS) rat is a well-characterized model of autosomal recessive retinitis pigmentosa (RP) due to a defect in the retinal pigment epithelium (RPE). It is homozygous for a null mutation in the gene encoding , a receptor tyrosine kinase found in RPE cells, that is required for phagocytosis of shed photoreceptor outer segments. The absence of Mertk results in accumulation of outer segment debris. This subsequently leads to progressive loss of photoreceptor cells. In order to evaluate the efficacy of lentiviral-mediated gene replacement therapy in the RCS rat, we produced recombinant VSV-G pseudotyped HIV-1-based lentiviruses containing a murine Mertk cDNA driven by a spleen focus forming virus (SFFV) promoter. The vector was subretinally injected into the right eye of 10-day-old RCS rats; the left eye was left untreated as an internal control. Here, we present a detailed assessment of the duration and extent of the morphological rescue and the resulting functional benefits. We examined animals at various time points over a period of 7 months by light and electron microscopy, and electroretinography. We observed correction of the phagocytic defect, slowing of photoreceptor cell loss and preservation of retinal function for up to 7 months. This study demonstrates the potential of gene therapy approaches for the treatment of retinal degenerations caused by defects specific to the RPE and supports the use of lentiviral vectors for the treatment of such disorders.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , HIV-1/genética , Epitélio Pigmentado Ocular/metabolismo , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Retinose Pigmentar/terapia , Animais , Eletrorretinografia , Humanos , Injeções , Microscopia Eletrônica , Modelos Animais , Células Fotorreceptoras/patologia , Epitélio Pigmentado Ocular/fisiopatologia , Epitélio Pigmentado Ocular/ultraestrutura , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Mutantes , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia , Vírus Formadores de Foco no Baço/genética , Fatores de Tempo , c-Mer Tirosina Quinase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...