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











Base de dados
Intervalo de ano de publicação
2.
Gene Ther ; 25(6): 450, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30046128

RESUMO

The authors originally published this article under the incorrect license type; this has now been corrected and is published under the CC-BY license.

3.
Gene Ther ; 24(12): 810-818, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29188796

RESUMO

The retinal pigment epithelium (RPE) interacts closely with photoreceptors to maintain visual function. In degenerative diseases such as Stargardt disease and age-related macular degeneration, the leading cause of blindness in the developed world, RPE cell loss is followed by photoreceptor cell death. RPE cells can proliferate under certain conditions, suggesting an intrinsic regenerative potential, but so far this has not been utilised therapeutically. Here, we used E2F2 to induce RPE cell replication and thereby regeneration. In both young and old (2 and 18 month) wildtype mice, subretinal injection of non-integrating lentiviral vector expressing E2F2 resulted in 47% of examined RPE cells becoming BrdU positive. E2F2 induced an increase in RPE cell density of 17% compared with control vector-treated and 14% compared with untreated eyes. We also tested this approach in an inducible transgenic mouse model of RPE loss, generated through activation of diphtheria toxin-A gene. E2F2 expression resulted in a 10-fold increase in BrdU uptake and a 34% increase in central RPE cell density. Although in mice this localised rescue is insufficiently large to be demonstrable by electroretinography, a measure of massed retinal function, these results provide proof-of-concept for a strategy to induce in situ regeneration of RPE for the treatment of RPE degeneration.


Assuntos
Fator de Transcrição E2F2/genética , Técnicas de Transferência de Genes , Terapia Genética , Degeneração Macular/terapia , Epitélio Pigmentado da Retina/fisiopatologia , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Proliferação de Células/genética , Toxina Diftérica/genética , Modelos Animais de Doenças , Vetores Genéticos , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Regeneração , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo
4.
Gene Ther ; 23(12): 857-862, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27653967

RESUMO

Leber congenital amaurosis is a group of inherited retinal dystrophies that cause severe sight impairment in childhood; RPE65-deficiency causes impaired rod photoreceptor function from birth and progressive impairment of cone photoreceptor function associated with retinal degeneration. In animal models of RPE65 deficiency, subretinal injection of recombinant adeno-associated virus (AAV) 2/2 vectors carrying RPE65 cDNA improves rod photoreceptor function, and intervention at an early stage of disease provides sustained benefit by protecting cone photoreceptors against retinal degeneration. In affected humans, administration of these vectors has resulted to date in relatively modest improvements in photoreceptor function, even when retinal degeneration is comparatively mild, and the duration of benefit is limited by progressive retinal degeneration. We conclude that the demand for RPE65 in humans is not fully met by current vectors, and predict that a more powerful vector will provide more durable benefit. With this aim we have modified the original AAV2/2 vector to generate AAV2/5-OPTIRPE65. The new configuration consists of an AAV vector serotype 5 carrying an optimized hRPE65 promoter and a codon-optimized hRPE65 gene. In mice, AAV2/5-OPTIRPE65 is at least 300-fold more potent than our original AAV2/2 vector.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Amaurose Congênita de Leber/terapia , cis-trans-Isomerases/genética , Células 3T3 , Animais , Feminino , Vetores Genéticos/administração & dosagem , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Coelhos , cis-trans-Isomerases/metabolismo
5.
Gene Ther ; 20(5): 545-55, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22951453

RESUMO

Recent clinical trials of retinal pigment epithelium gene (RPE65) supplementation therapy in Leber congenital amaurosis type 2 patients have demonstrated improvements in rod and cone function, but it may be some years before the effects of therapy on photoreceptor survival become apparent. The Rpe65-deficient dog is a very useful pre-clinical model in which to test efficacy of therapies, because the dog has a retina with a high degree of similarity to that of humans. In this study, we evaluated the effect of RPE65 gene therapy on photoreceptor survival in order to predict the potential benefit and limitations of therapy in patients. We examined the retinas of Rpe65-deficient dogs after RPE65 gene therapy to evaluate the preservation of rods and cone photoreceptor subtypes. We found that gene therapy preserves both rods and cones. While the moderate loss of rods in the Rpe65-deficient dog retina is slowed by gene therapy, S-cones are lost extensively and gene therapy can prevent that loss, although only within the treated area. Although LM-cones are not lost extensively, cone opsin mislocalization indicates that they are stressed, and this can be partially reversed by gene therapy. Our results suggest that gene therapy may be able to slow cone degeneration in patients if intervention is sufficiently early and also that it is probably important to treat the macula in order to preserve central function.


Assuntos
Amaurose Congênita de Leber/terapia , Células Fotorreceptoras Retinianas Cones , Células Fotorreceptoras Retinianas Bastonetes , cis-trans-Isomerases/genética , Animais , Sobrevivência Celular/genética , Modelos Animais de Doenças , Cães , Terapia Genética , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/patologia , Retina/efeitos dos fármacos , Retina/patologia , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Células Fotorreceptoras Retinianas Bastonetes/patologia , cis-trans-Isomerases/administração & dosagem , cis-trans-Isomerases/deficiência
6.
Gene Ther ; 19(2): 154-61, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22033465

RESUMO

Over the last decade, gene supplementation therapy for inherited retinal degeneration has come of age. Early proof-of-concept studies in animal models of disease showed modest, but genuine improvements in retinal function and/or survival. Further development of the vectors used for gene transfer to the retina has led to better treatment efficacy in a wide variety of animal models, leading in 2008 to the initiation of three clinical trials for Leber congenital amaurosis caused by retinal pigment epithelium 65 deficiency. The results from these trials suggest that the treatment of inherited retinal dystrophy by gene therapy can be safe and effective. Here, we examine the progress of gene supplementation therapy in the retina, and discuss the potential for using gene therapy to treat different forms of inherited retinal degeneration.


Assuntos
Terapia Genética , Amaurose Congênita de Leber/terapia , Retina/patologia , Distrofias Retinianas/genética , Distrofias Retinianas/terapia , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Genes Recessivos , Humanos , Amaurose Congênita de Leber/genética , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia
7.
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
8.
Gene Ther ; 17(4): 486-93, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20010626

RESUMO

Gene therapy for inherited retinal degeneration in which expression of a mutant allele has a gain-of-function effect on photoreceptor cells is likely to depend on efficient silencing of the mutated allele. Peripherin-2 (Prph2, also known as peripherin/RDS) is an abundantly expressed photoreceptor-specific gene. In humans, gain-of-function mutations in PRPH2 result in both autosomal dominant retinitis pigmentosa and dominant maculopathies. Gene-silencing strategies for these conditions include RNA interference by short hairpin RNAs (shRNAs). Recent evidence suggests that microRNA (miRNA)-based hairpins may offer a safer and more effective alternative. In this study, we used for the first time a virally transferred miRNA-based hairpin to silence Prph2 in the murine retina. The results show that an miRNA-based shRNA can efficiently and specifically silence Prph2 in vivo as early as 3 weeks after AAV2/8-mediated subretinal delivery, leading to a nearly 50% reduction of photoreceptor cells after 5 weeks. We conclude that miRNA-based hairpins can achieve rapid and robust gene silencing after efficient vector-mediated delivery to the retina. The rationale of using an miRNA-based template to improve the silencing efficiency of a hairpin may prove valuable for allele-specific silencing in which the choice for an RNAi target is limited and offers an alternative therapeutic strategy for the treatment of dominant retinopathies.


Assuntos
Terapia Genética/métodos , Proteínas de Filamentos Intermediários/genética , Glicoproteínas de Membrana/genética , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Interferência de RNA , Degeneração Retiniana/terapia , Animais , Pareamento de Bases , Sequência de Bases , Western Blotting , Primers do DNA/genética , Dependovirus , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Periferinas , Degeneração Retiniana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Eye (Lond) ; 23(10): 1898-903, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19151647

RESUMO

Gene-based therapies offer the means to address gene defects responsible for inherited retinal disorders. A number of studies in experimental and preclinical models have demonstrated proof-of-principle that gene replacement therapy can mediate significant quantifiable improvements in ocular morphology and visual function. The first results of clinical trials of gene therapy for early-onset severe retinal dystrophy caused by defects in RPE65 show proof-of-concept for efficacy and short-term safety in humans. The challenges for gene therapy of conditions caused by gain-of-abnormal function are being addressed by strategies to knock down expression of the disease allele. Vector-mediated expression of neuroprotective proteins may offer a generic approach for preserving vision in single-gene and multi-gene retinal degenerations. Gene therapy is likely to be most successful where stable expression of the therapeutic transgene can be achieved at an appropriate level in diseases in which retinal development is unaffected and a significant number of target cells survive at the point of intervention.


Assuntos
Terapia Genética/métodos , Doenças Retinianas/genética , Doenças Retinianas/terapia , Animais , Proteínas de Transporte/genética , Modelos Animais de Doenças , Proteínas do Olho/genética , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Mutação , Doenças Retinianas/congênito , cis-trans-Isomerases
10.
Gene Ther ; 15(22): 1478-88, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18580969

RESUMO

Uveitis is a sight threatening inflammatory disorder that remains a significant cause of visual loss. We investigated lentiviral gene delivery of interleukin 1 receptor antagonist (IL-1ra) or interleukin (IL)-10 to ameliorate murine endotoxin-induced uveitis (EIU). An human immunodeficiency virus-1-based vector containing the mIL-1ra or mIL-10 cDNA demonstrated high expression of biologically active cytokine. Following administration of Lenti.GFP into the anterior chamber, transgene expression was observed in corneal endothelial cells, trabecular meshwork and iris cells. To treat EIU, mice were injected with Lenti.IL-1ra, Lenti.IL-10 or a combination of these. EIU was induced 14 days after vector administration and mice were culled 12 h following disease induction. Lenti.IL-1ra or Lenti.IL-10-treated eyes showed significantly lower mean inflammatory cell counts in the anterior and posterior chambers compared with controls. The aqueous total protein content was also significantly lower in treated eyes, demonstrating better preservation of the blood-ocular barrier. Furthermore, the treated eyes showed less in vivo fluorescein leakage from inner retinal vessels compared with controls. The combination of both IL-1ra and IL-10 had no additive effect. Thus, lentiviral gene delivery of IL-1ra or IL-10 significantly reduces the severity of experimental uveitis, suggesting that lentiviral-mediated expression of immunomodulatory genes in the anterior chamber offers an opportunity to treat uveitis.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , HIV-1/genética , Proteína Antagonista do Receptor de Interleucina 1/genética , Interleucina-10/genética , Uveíte/terapia , Animais , Feminino , Expressão Gênica , Vetores Genéticos/genética , Humanos , Injeções , Proteína Antagonista do Receptor de Interleucina 1/imunologia , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Transdução Genética/métodos , Transgenes , Úvea/imunologia , Uveíte/imunologia
11.
Gene Ther ; 13(16): 1191-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16838031

RESUMO

The eye has unique advantages as a target organ for gene therapy of both inherited and acquired ocular disorders and offers a valuable model system for gene therapy. The eye is readily accessible to phenotypic examination and investigation of therapeutic effects in vivo by fundus imaging and electrophysiological techniques. Considerable progress has been made in the development of gene replacement therapies for retinal degenerations resulting from gene defects in photoreceptor cells (rds, RPGRIP, RS-1) and in retinal pigment epithelial cells (MerTK, RPE65, OA1) using recombinant adeno-associated virus and lentivirus-based vectors. Gene therapy also offers a potentially powerful approach to the treatment of complex acquired disorders such as those involving angiogenesis, inflammation and degeneration, by the targeted sustained intraocular delivery of therapeutic proteins. Proposals for clinical trials of gene therapy for early-onset retinal degeneration owing to defects in the gene encoding the visual cycle protein RPE65 have recently received ethical approval.


Assuntos
Oftalmopatias/terapia , Terapia Genética/tendências , Ensaios Clínicos como Assunto , Dependovirus/genética , Oftalmopatias/metabolismo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Engenharia Genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Retina/metabolismo
12.
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
13.
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
14.
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
15.
Mol Cell Neurosci ; 25(1): 103-10, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14962744

RESUMO

Inherited retinal degenerations are a major cause of blindness for which there are currently no effective therapies. Significant progress concerning in vivo gene transfer has allowed retardation of degeneration or retinal functional improvement in different animal models. To date, there has been no evaluation of the impact of these treatments on higher visual function, a critical step for validating gene therapy treatment strategies. Here, we have used adeno-associated (AAV2)-mediated gene transfer of Prph2 in the Prph2(Rd2/Rd2) mouse model. We then assessed higher visual function by recording from central visually responsive neurons in the superior colliculus and improvements were correlated in individual animals with retinal function (ERG) and histological and biochemical changes. Although gene replacement therapy only partially restores photoreceptor morphology, it results in a 300% increase of the visual cycle protein rhodopsin, leading to retinal function improvement (250% increase of b-wave amplitude) and significantly higher central visual responses (166% increase at 24 cd/m(2)). These findings suggest that gene replacement therapy leading to even relatively modest structural improvement may result in improved central visual function.


Assuntos
Terapia Genética/métodos , Proteínas de Filamentos Intermediários/deficiência , Glicoproteínas de Membrana/deficiência , Regeneração Nervosa/genética , Proteínas do Tecido Nervoso/deficiência , Degeneração Retiniana/terapia , Colículos Superiores/crescimento & desenvolvimento , Vias Visuais/crescimento & desenvolvimento , Potenciais de Ação/genética , Animais , Modelos Animais de Doenças , Eletrorretinografia , Terapia Genética/tendências , Vetores Genéticos/genética , Proteínas de Filamentos Intermediários/genética , Glicoproteínas de Membrana/genética , Potenciais da Membrana/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Periferinas , Estimulação Luminosa , Células Fotorreceptoras/crescimento & desenvolvimento , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Recuperação de Função Fisiológica/genética , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Rodopsina/metabolismo , Colículos Superiores/citologia , Colículos Superiores/metabolismo , Transmissão Sináptica/genética , Regulação para Cima/genética , Visão Ocular/genética , Vias Visuais/citologia , Vias Visuais/metabolismo
16.
Gene Ther ; 10(16): 1336-44, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12883530

RESUMO

Recombinant adeno-associated virus (rAAV) vectors are attractive candidates for the treatment of inherited and acquired retinal disease. Although rAAV vectors are well characterized in rodent models, a prerequisite to their clinical application in human patients is the thorough evaluation of their efficacy and safety in intermediate animal models. In this study, we describe rAAV-2-mediated expression of GFP reporter gene in retinal cells following local vector delivery in dogs. Subretinal delivery of rAAV.CMV.GFP was performed unilaterally in eight normal dogs from 6 weeks of age. The area of retinal transduction was maximized by the optimization of surgical techniques for subretinal vector delivery by pars-plana vitrectomy and the use of fine-gauge subretinal cannulae to create multiple retinotomies. rAAV-2 vectors mediated efficient stable reporter gene expression in photoreceptors and retinal pigment epithelial cells. We found efficient transduction of cone photoreceptors in addition to rods in both the canine retina and after subretinal vector delivery in another intermediate animal model, the feline retina. GFP expression in dogs was confined to the area of the retinal bleb and was sustained in cells at this site for at least 18 months. Electroretinography demonstrated a modest reduction in global rod-mediated retinal function following subretinal delivery of rAAV.CMV.GFP. Three of the eight animals developed delayed-onset intraocular inflammation, in two cases associated with a serum antibody response to GFP protein. We conclude that rAAV-2 vectors mediate efficient sustained transgene expression in rod and cone photoreceptors following subretinal delivery in this intermediate animal model. The possibility of adverse effects including intraocular immune responses and reduced retinal function requires further investigation prior to clinical applications in patients.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Células Fotorreceptoras de Vertebrados/metabolismo , Doenças Retinianas/terapia , Transdução Genética/métodos , Animais , Células Cultivadas , Cães , Eletrorretinografia , Fundo de Olho , Expressão Gênica , Proteínas de Fluorescência Verde , Inflamação , Proteínas Luminescentes/genética , Células Fotorreceptoras de Vertebrados/imunologia , Células Fotorreceptoras de Vertebrados/fisiologia , Epitélio Pigmentado Ocular/metabolismo , Fatores de Tempo
17.
Gene Ther ; 10(12): 1049-54, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12776163

RESUMO

Recombinant AAV vectors mediate efficient and sustained transgene expression in retinal tissues and offer a powerful approach to the local, sustained delivery of angiostatic proteins for the treatment of ocular neovascular disorders. The application of such strategies may also require regulated gene expression to minimize the potential for unwanted adverse effects. In this study, we have evaluated the effect of a hypoxia-responsive element (HRE) on the kinetics of recombinant adeno-associated (rAAV)-mediated reporter gene expression in murine models of retinal and choroidal neovascularization. In murine ischaemia-induced retinal neovascularization, intravitreal delivery of rAAV.HRE.GFP results in reporter gene expression specifically at sites of vascular closure during the period of active neovascularization and not after vector delivery in normal controls. In murine laser-induced choroidal neovascularization, subretinal delivery of rAAV.HRE.GFP results in reporter gene expression at sites of active neovascularization but not elsewhere or after vector delivery in normal controls. HRE-driven gene expression offers an attractive strategy for the targeted and regulated delivery of angiostatic proteins to the retina in the management of neovascular disorders.


Assuntos
Neovascularização de Coroide/terapia , Regulação da Expressão Gênica , Marcação de Genes/métodos , Terapia Genética/métodos , Neovascularização Retiniana/terapia , Animais , Hipóxia Celular/genética , Dependovirus/genética , Técnicas de Transferência de Genes , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde , Hipóxia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Elementos de Resposta/genética , Transgenes
18.
Gene Ther ; 10(6): 523-7, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12621456

RESUMO

Intraocular delivery of a variety of neurotrophic factors has been widely investigated as a potential treatment for retinal dystrophy (RD). The most commonly studied factor, ciliary neurotrophic factor (CNTF), has been shown to preserve retinal morphology and to promote cell survival in a variety of models of RD. In order to evaluate CNTF as a potential treatment for RD, we used the Prph2(Rd2/Rd2) mouse. CNTF was expressed intraocularly using AAV-mediated gene delivery either by itself or, in a second treatment group, combined with AAV-mediated gene replacement therapy of peripherin2, which we have previously shown to improve photoreceptor structure and function. We confirmed in both groups of animals that CNTF reduces the loss of photoreceptor cells. Visual function, however, as assessed over a time course by electroretinography (ERG), was significantly reduced compared with untreated controls. Furthermore, CNTF gene expression negated the effects on function of gene replacement therapy. In order to test whether this deleterious effect is only seen when degenerating retina is treated, we recorded ERGs from wild-type mice following intraocular injection of AAV expressing CNTF. Here a marked deleterious effect was noted, in which the b-wave amplitude was reduced by at least 50%. Our results demonstrate that intraocular CNTF gene delivery may have a deleterious effect on the retina and caution against its application in clinical trials.


Assuntos
Fator Neurotrófico Ciliar/genética , Terapia Genética/efeitos adversos , Glicoproteínas de Membrana , Retina/fisiopatologia , Degeneração Retiniana/terapia , Transdução Genética/métodos , Animais , Sobrevivência Celular , Dependovirus/genética , Eletrorretinografia , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Injeções , Proteínas de Filamentos Intermediários/genética , Camundongos , Camundongos Endogâmicos , Modelos Animais , Proteínas do Tecido Nervoso/genética , Periferinas , Células Fotorreceptoras de Vertebrados/patologia , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia
19.
Gene Ther ; 9(5): 320-6, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11938451

RESUMO

Retinal angiogenesis is a central feature of the leading causes of blindness. Current treatments for these conditions are of limited efficacy and cause significant adverse effects. In this study, we evaluated the angiostatic effect of gene transfer of the soluble VEGF receptor sFlt-1 in a mouse model of ischaemia-induced retinal neovascularisation using adenovirus and adeno-associated virus (AAV) vectors. We induced proliferative retinopathy in mice by exposure to 75% oxygen from postnatal day 7 (p7) to p12 and injected intravitreally recombinant viral vectors expressing the reporter green fluorescent protein (GFP) or vectors expressing the VEGF inhibitor sFlt-1. Efficient adenovirus-mediated GFP expression was evident in cells of the corneal endothelium and iris pigment epithelium. AAV-mediated GFP expression was evident in ganglion cells and cells of the inner nuclear layer of the retina. Vector-mediated sFlt-1 expression was confirmed by ELISA of pooled homogenised whole eyes. Injection of either vector expressing sFlt-1 resulted in a reduction in the number of neovascular endothelial cells by 56% and 52% for adenovirus and AAV vectors, respectively (P < 0.05). Local gene transfer of sFlt-1 consistently inhibits experimental retinal neovascularisation by approximately 50% and offers a powerful novel approach to the clinical management of retinal neovascular disorders.


Assuntos
Proteínas da Matriz Extracelular/genética , Terapia Genética/métodos , Neovascularização Retiniana/terapia , Adenoviridae/genética , Animais , Dependovirus/genética , Expressão Gênica , Vetores Genéticos/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Cadeias Pesadas de Miosina , Miosina não Muscular Tipo IIB
20.
Vision Res ; 42(4): 541-9, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11853771

RESUMO

Using confocal microscopy we have examined in detail the temporal and spatial pattern of green fluorescent protein expression following sub-retinal injection of recombinant adeno-associated virus (rAAV) in the mouse and have determined the effect of viral titre on the number and type of cells transduced. Our results suggest that some transgene expression occurs as early as three days after injection, and that transgene expression occurs beyond the area of retinal detachment. Vector titre appears to have a substantial effect on both transduction efficiency and the speed of onset of photoreceptor cell transduction. Our data suggests that we have not yet reached the limits of photoreceptor transduction efficiency using AAV vectors. An increase in titre could still lead to an improved transduction efficiency and faster onset of photoreceptor transduction. We failed to detect transfected cones even in areas where nearly 100% of the rods were transduced, but we found efficient and sustained RPE transduction.


Assuntos
Terapia Genética/métodos , Modelos Animais , Retina/metabolismo , Retinose Pigmentar/terapia , Transfecção/métodos , Transgenes , Animais , Citomegalovirus/genética , Dependovirus/genética , Expressão Gênica , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde , Injeções , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos CBA , Epitélio Pigmentado Ocular/metabolismo , Distribuição Aleatória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA