Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Med (Berl) ; 96(10): 1107-1118, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30105447

RESUMO

Aberrant growth of blood vessels in the choroid layer of the eye, termed choroidal neovascularization (CNV), is the pathological hallmark of exudative age-related macular degeneration (AMD), causing irreversible blindness among the elderly. Co-localization of proangiogenic factors and hypoxia inducible factors (HIF) in neovascular membranes from AMD eyes suggests the role of hypoxia in pathogenesis of CNV. In order to utilize hypoxic conditions in RPE for therapeutic purposes, we developed an optimized hypoxia regulated, RPE cell-specific gene therapy to inhibit choroidal neovascularization. An adeno-associated virus (AAV2) vector comprising a RPE-specific promoter and HIF-1 response elements (HRE) was designed to regulate production of human endostatin (a powerful angiostatic protein) in RPE. The vector was tested in a mouse model of laser-induced CNV using subretinal delivery. Spectral domain optical coherence tomography (SD-OCT) images from live mice and confocal images from lectin stained RPE flat mount sections demonstrated reduction in CNV areas by 80% compared to untreated eyes. Quantitative real-time polymerase chain reaction (qPCR) confirmed exogenous endostatin mRNA expression from the regulated vector that was significantly elevated 3, 7, and 14 days following laser treatment, but its expression was completely shut off after 45 days. Thus, RPE-specific, hypoxia-regulated delivery of anti-angiogenic proteins could be a valuable therapeutic approach to treat neovascular AMD at the time and in the ocular space where it arises. KEY POINTS: An optimized gene therapy vector targeting hypoxia and tissue-specific expression has been designed. The inhibitory role of gene therapy vector was tested in a mouse model of laser-induced CNV. An 80% reduction in choroidal neovascularization was achieved by the optimized vector. The expression of endostatin was limited to retinal pigment epithelium and regulated by hypoxia.


Assuntos
Neovascularização de Coroide/terapia , Terapia Genética , Hipóxia , Animais , Dependovirus , Endostatinas/genética , Endostatinas/metabolismo , Vetores Genéticos , Camundongos Endogâmicos C57BL , Parvovirinae/genética , Epitélio Pigmentado da Retina/metabolismo
2.
Invest Ophthalmol Vis Sci ; 55(12): 8044-53, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25377223

RESUMO

PURPOSE: Müller cells, the major glial cell in the retina, play a significant role in retinal neovascularization in response to tissue hypoxia. We previously designed and tested a vector using a hypoxia-responsive domain and a glial fibrillary acidic protein (GFAP) promoter to drive green fluorescent protein (GFP) expression in Müller cells in the murine model of oxygen-induced retinopathy (OIR). This study compares the efficacy of regulated and unregulated Müller cell delivery of endostatin in preventing neovascularization in the OIR model. METHODS: Endostatin cDNA was cloned into plasmids with hypoxia-regulated GFAP or unregulated GFAP promoters, and packaged into self-complementary adeno-associated virus serotype 2 vectors (scAAV2). Before placement in hyperoxia on postnatal day (P)7, mice were given intravitreal injections of regulated or unregulated scAAV2, capsid, or PBS. Five days after return to room air, on P17, neovascular and avascular areas, as well as expression of the transgene and vascular endothelial growth factor (VEGF), were compared in OIR animals treated with a vector, capsid, or PBS. RESULTS: The hypoxia-regulated, glial-specific, vector-expressing endostatin reduced neovascularization by 93% and reduced the central vaso-obliteration area by 90%, matching the results with the unregulated GFAP-Endo vector. Retinas treated with the regulated endostatin vector expressed substantial amounts of endostatin protein, and significantly reduced VEGF protein. Endostatin production from the regulated vector was undetectable in retinas with undamaged vasculature. CONCLUSIONS: These findings suggest that the hypoxia-regulated, glial cell-specific vector expressing endostatin may be useful for treatment of neovascularization in proliferative diabetic retinopathy.


Assuntos
Hipóxia Celular/fisiologia , Retinopatia Diabética/terapia , Terapia Genética/métodos , Neuroglia/fisiologia , Neovascularização Retiniana/terapia , Animais , DNA Complementar , Retinopatia Diabética/fisiopatologia , Modelos Animais de Doenças , Endostatinas/metabolismo , Inativação Gênica/efeitos dos fármacos , Vetores Genéticos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Neovascularização Retiniana/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Invest Ophthalmol Vis Sci ; 52(12): 8562-70, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21960554

RESUMO

PURPOSE: Retinal Müller cells span the retina and secrete several trophic factors and represent the functional link between blood vessels and neurons, making them attractive targets for gene therapy. Therefore, a hypoxia-regulated, retinal glial cell-specific vector was constructed and tested for its response to hypoxia. METHODS: A hybrid promoter containing domains of human glial fibrillary acidic protein (GFAP) and several hypoxia-responsive and aerobically silenced elements (HRSE) was incorporated separately into plasmid vectors for generation of self-complementary adeno-associated virus. Müller cells trasfected with plasmids or virus were compared with other cell lines using standard METHODS: The mouse model of oxygen-induced retinopathy (OIR) was used to analyze retinas from mice exposed to high oxygen or room air to evaluate the induction of the regulated promoter. RESULTS: The regulated promoter was silenced under aerobic conditions in comparison with unregulated promoter in Müller cells. Hypoxia induced a 12-fold and 16-fold increase in promoter activity in primary Müller cells and human Müller cell lines, respectively. In the OIR model, intravitreal injection of the regulated promoter at postnatal day 7 (P7) resulted in high levels of green fluorescent protein expression only in retinal Müller cells at P17. GFP expression was absent in retinas of mice only exposed to room air. In vivo studies confirm normoxia silencing, hypoxic induction, and cell specificity of the regulated promoter in the mouse retina. CONCLUSIONS: This hypoxia-regulated, retinal glial cell-specific AAV vector provides a platform for gene therapy within regions of retinal hypoxia which are found in diabetic retinopathy and age-related macular degeneration.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/genética , Proteína Glial Fibrilar Ácida/genética , Hipóxia/terapia , Neuroglia/fisiologia , Doenças Retinianas/terapia , Adenoviridae/genética , Animais , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Inativação Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Hipóxia/fisiopatologia , Camundongos , Neuroglia/citologia , Oxigênio/farmacologia , Plasmídeos/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Retina/citologia , Retina/fisiologia , Doenças Retinianas/fisiopatologia
4.
Invest Ophthalmol Vis Sci ; 49(7): 3207-15, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18579766

RESUMO

PURPOSE: Metallothioneins (MTs) in the brain and retina are believed to bind metals and reduce free radicals, thereby protecting neurons from oxidative damage. This study was undertaken to investigate whether retinal photoreceptor (PR) cells lacking MTs are more susceptible to hyperbaric oxygen (HBO)-induced cell death in vivo. METHODS: Wild-type (WT) and MT-knockout (MT-KO) mice lacking metallothionein (MT)-1 and MT-2 were exposed to three atmospheres of 100% oxygen for 3 hours, 3 times per week for 1, 3, or 5 weeks. The control animals were not exposed. Histologic analysis of PR viability was performed by counting rows of nuclei in the outer nuclear layer (ONL). Ultrastructure studies verified PR damage. RESULTS: HBO exposure produced a major loss of PR cells in the central retinas of WT and MT-KO mice, with no effect on the peripheral retina even at the longest (5 weeks) exposures. The degree of PR damage and cell death increased with duration of HBO exposure. One week of HBO exposure was insufficient to cause PR death, but tissue damage was observed in the inner and outer segments. At 3 weeks, the rows of PR nuclei in the central retina were significantly reduced by 38% in WT and 28% in MT-KO animals. At 5 weeks, PR loss was identical in WT (34%) and MT-KO (34%) animals and was comparable to that in WT at 3 weeks. CONCLUSIONS: The data suggest that MT-1 and -2 alone are not sufficient for protecting PRs against HBO-induced cell death. The selective degeneration of central PRs may provide clues to mechanisms of oxidative damage in retinal disease.


Assuntos
Oxigenoterapia Hiperbárica/efeitos adversos , Metalotioneína/deficiência , Células Fotorreceptoras , Degeneração Retiniana/etiologia , Degeneração Retiniana/metabolismo , Animais , Morte Celular , Núcleo Celular/patologia , Sobrevivência Celular , Suscetibilidade a Doenças , Camundongos , Camundongos Knockout , Células Fotorreceptoras/patologia , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia
5.
Mol Vis ; 14: 125-35, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18334927

RESUMO

PURPOSE: High oxygen consumption and cyclical changes related to dark-adaptation are characteristic of the outer retina. Oxygenation changes may contribute to the selective vulnerability of the retina in retinitis pigmentosa (RP) patients, especially for those forms involving genes with global cellular functions. Genes coding for components of the U4/U6.U5 tri small nuclear ribonucleoprotein (tri-snRNP) complex of the spliceosome stand out, because mutations in four genes cause RP, i.e., RP9 (PAP1), RP11 (PRPF31), RP13 (PRPF8), and RP18 (PRPF3), while there is no degeneration outside the retina despite global expression of these genes. With the assumption that variable oxygenation plays a role in RP forms related to pre-mRNA splicing and the retina and brain are similar, we searched a data collection of ischemia-hypoxia regulated genes of the brain for oxygen regulated genes of the U4/U6.U5 tri-snRNP complex. METHODS: A database of ischemia-hypoxia response (IHR) genes in the brain was generated from gene expression profiling studies [n=24]. Public databases (NCBI) were searched for RP genes with global function that are expressed in the brain. From the IHR gene list, we extracted genes that were directly related to retinal degeneration through a listed mutation (OMIM, Retnet, RISN). The database was then examined for indirect links to RP forms affecting the U4/U6.U5 tri-snRNP complex by searching for IHR genes contributing to this complex. Potential expression of matched genes in the retina was ascertained using NEIBank. Immunohistochemistry was used to localize a selected protein of the U4/U6.U5 tri-snRNP complex in cynomolgus monkey and human retina specimens. RESULTS: The approach identified genes that cause retinal degeneration (CNGB1, SEMA4A, RRG4) or developmental changes (SOX2) when mutated. One IHR gene, Pim1, is the immediate binding partner for PAP1 (RP9). Three IHR genes linked the U4/U6.U5 tri-snRNP complex to regulation by oxygenation: PRPF4; SART1, also known as 110 kDa SR-related protein of the U4/U6.U5 tri-snRNP or as hypoxia associated factor (HAF); and LSM8, U6 snRNA-associated Sm-like protein. The 110 kDa SR-related protein was localized in all retinal cells including photoreceptors. CONCLUSIONS: Regulation by changes in oxygenation within the U4/U6.U5 tri-snRP complex could be particularly important for photoreceptors where oxygen consumption follows a circadian rhythm. If the U4/U6.U5 tri-snRP complex is already impaired by mutations in any of the four genes causing RP, it may be unable to follow properly the physiological demands of oxygenation which are mediated by the four hypoxia-regulated proteins emerging in this study. Selective vulnerability may involve complex combinations of widely expressed genes, specific cellular functions and local energy availability.


Assuntos
Genes Dominantes , Hipóxia Encefálica/genética , Retinose Pigmentar/genética , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Processamento Alternativo/genética , Animais , Antígenos de Neoplasias/genética , Isquemia Encefálica/genética , Bases de Dados Genéticas , Cães , Humanos , Hipóxia Encefálica/metabolismo , Imuno-Histoquímica , Macaca fascicularis , Masculino , Camundongos , Estresse Oxidativo , Proteínas Associadas a Pancreatite , Retinose Pigmentar/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/genética , Ribonucleoproteína Nuclear Pequena U5/genética , Ribonucleoproteínas Nucleares Pequenas/genética
6.
Mol Vis ; 14: 471-80, 2008 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-18334957

RESUMO

PURPOSE: To develop an hypoxia-regulated retinal pigment epithelium (RPE)-specific adeno-associated virus (AAV) gene transfer platform that exploits hypoxia as a physiologic trigger for an early antiangiogenic treatment strategy directed at arresting neovascularization in the eye. METHODS: Tissue-specific and hypoxia-regulated expression vectors were constructed with tandem combinations of hypoxia responsive elements and aerobically silenced elements (HRSE) that together induce gene expression in hypoxia and suppress it in normoxia. For RPE-specific expression, the HRSE and a (6x) HRE (hypoxia responsive element) oligomer were ligated upstream of the Rpe65 promoter in a pGL3 firefly luciferase plasmid (pGL3-HRSE-6xHRE-Rpe65). The cell specificity of this novel hybrid promoter was tested in human RPE (ARPE-19), human glioblastoma, rat C6 glioma, mouse hippocampal neurons, and human embryonic kidney cell lines. Expression of all cell types in normoxia, and following 40 h hypoxia, was analyzed by dual luciferase assay. After confirmation of its tissue-specificity and hypoxia-inducibility, the hybrid promoter construct was integrated into a replication-deficient AAV delivery system and tested for cell-selectivity and hypoxia-inducible green fluorescent protein (GFP) reporter expression. RESULTS: The HRSE-6xHRE-Rpe65 promoter was highly selective for RPE cells, strongly induced in hypoxia, and similar in activation strength to the cytomegalovirus (CMV) promoter. The AAV.HRSE.6xHRE.Rpe65 vector induced robust GFP expression in hypoxic ARPE-19 cells, but elicited no GFP expression in hypoxia in other cell types or in normoxic ARPE-19 cells. CONCLUSIONS: The hypoxia-regulated, aerobically-silenced RPE-targeting vector forms a platform for focal autoregulated delivery of antiangiogenic agents in hypoxic regions of the RPE. Such autoinitiated therapy provides a means for early intervention in choroidal neovascularization, when it is most sensitive to inhibition.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Epitélio Pigmentado Ocular/virologia , Transdução Genética/métodos , Animais , Hipóxia Celular , Linhagem Celular , Proteínas do Olho/genética , Inativação Gênica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Luciferases/metabolismo , Camundongos , Especificidade de Órgãos , Elementos de Resposta/genética , Transfecção
7.
Mol Vis ; 12: 756-67, 2006 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-16862069

RESUMO

PURPOSE: To compare the transduction efficiency of a lentiviral vector in the retina of normal mice and retinal degenerative (rd) mice following subretinal injection at various postnatal ages. METHODS: Subretinal injections of lentiviral vector (pHR-CMV-GFP, 107IU/ml) were performed in normal (C57/6J) and rd mice on postnatal days P1 to P7 using a trans-scleral method and on days P14-P35 by a trans-corneal method. One to six weeks later the eyes were prepared for histological analysis. GFP positive cells were identified in retinal sections and retinal whole mounts to determine the overall extent and distribution of lentiviral transduction. RESULTS: Expression of GFP was observed adjacent to the injection site starting about 1 week after injection in both normal and rd mice and lasted 6 weeks (the longest period examined). In normal mice, GFP expression continued to increase and peaked around 2-3 weeks after injection with expression varying from approximately one quarter to the entire retina. GFP expression peaked earlier in rd mice injected from P1 to P7 compared to normal mice. Lenti-GFP expression decreased rapidly in rd mice older than P15. This was attributed to a period of intensive photoreceptor (PR) degeneration characteristic to this mutant. Retinal GFP expression was virtually absent in eyes injected after P14 in both normal and rd mice. Histological sections from P3 injected eyes showed GFP expression 9 days post-injection in both retinal pigment epithelium (RPE) and photoreceptor (PR) cells. GFP expression in RPE cells was stronger than that in PR cells. Both rods and cones expressed the lenti-GFP. GFP expression was limited to the RPE of normal mice if injections were performed at P14 or later. In rd mice, GFP expression in RPE was observed one week after injection at P1; GFP+-PR and -RPE cells were first detected 9 days after injection at P1, and 7 days after injection at P3-P7; RPE cells and occasional Muller cells around the injection site were GFP+ when the injection was performed at P14 or later. CONCLUSIONS: Lentiviral-mediated GFP transduction of RPE was efficient and sustained at all ages examined in both the normal and rd mouse. Trans-scleral, subretinal injection of lenti-GFP during the first postnatal week produced age-dependent transduction of PR cells in both mouse strains. Lenti-GFP expression was absent in both mouse strains if injections occurred after P14. There was a dramatic decrease in the transduction efficiency in rd mouse retinas corresponding to the degeneration of PR cells. However, the early stages of retinal degeneration in rd mice appeared to increase the transduction efficiency of PR cells. These data suggest that both age and degree of PR degeneration are important parameters to consider when designing gene therapy experiments or protocols.


Assuntos
Lentivirus/genética , Degeneração Retiniana/genética , Transdução Genética , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Injeções , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/patologia
8.
Exp Eye Res ; 79(2): 189-201, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15325566

RESUMO

Naturally occurring mutations of the beta subunit of the cyclic guanosine monophosphate (cGMP) phosphodiesterase (beta-PDE) gene in rod photoreceptors of mice and dogs are similar to one of the inherited retinal degenerations termed retinitis pigmentosa in humans. Defects in the rod beta-PDE gene leading to photoreceptor cell degeneration in retinal degenerative (rd) mice can be corrected by transfer of a wild type beta-PDE gene. However, the rapid photoreceptor degeneration in this mutant makes the study of gene therapy difficult. Since the retinal degeneration is slowed in vitro, we have employed retinal explants from rd mice to study factors influencing viral transduction. Retinal explants provide a rapid, efficient method to compare the transduction efficiency of adenoviral vector-mediated reporter gene delivery at different ages in normal and rd mice. Retinal explants from postnatal day (P)2 to P28 control (C57BL/6J) and P2-P42 rd mice were exposed for 20 hr to 2.5 x 10(8) plaque forming units (pfu) ml(-1) of adenoviral vector with a beta-galactosidase (Lac Z) reporter gene (Ad-CMV-Lac Z). After incubation in vector-free media for an additional 3 days, the explants were fixed and histochemically stained for beta-galactosidase to reveal Lac Z gene expression. The explants were also embedded and sectioned for light microscopic observation. Transduction efficiency was higher in rd mice than in controls on all postnatal days examined. In normal retinal explants, expression of the Lac Z gene increased from P2 to a peak around P7-P8, then decreased at subsequent ages; little transduction could be found after P17. In rd mice transduction efficiency of Ad-CMV-Lac Z increased from P2 to P7, decreased by P10 and increased again after P10. The most dramatic increase in the transduction efficiency occurred in the rd retina between P10 and P15 when Lac Z was intensely expressed throughout the retina. Microscopic examination of retinal sections revealed the types and distribution of Lac Z-positive cells responsible for the deep blue staining in the retinal whole mount. In normal and rd mice, Lac Z-positive cells were located throughout the retina. However, larger numbers of Lac Z-positive cells were present at all ages examined in retinal explants from rd mice compared to normal mice. These data indicate a difference in transduction efficiency between normal and rd mice, especially after P12, and suggest efficient adenovirus-mediated gene transfer is more attainable in developing or degenerating retina. Thus, transduction efficiency in rd mice depends on the relationship between development, maturation and the degenerative state of the photoreceptor cells.


Assuntos
3',5'-GMP Cíclico Fosfodiesterases/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Degeneração Retiniana/terapia , 3',5'-GMP Cíclico Fosfodiesterases/metabolismo , Adenoviridae/genética , Fatores Etários , Animais , Vetores Genéticos , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Retina/enzimologia , Retina/crescimento & desenvolvimento , Degeneração Retiniana/enzimologia , Degeneração Retiniana/patologia , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...