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.
Biomed Opt Express ; 10(1): 151-166, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30775090

RESUMO

In cancer research there is a fundamental need for animal models that allow the in vivo longitudinal visualization and quantification of tumor development, nanotherapeutic delivery, the tumor microenvironment including blood vessels, macrophages, fibroblasts, immune cells, and extracellular matrix, and the tissue response to treatment. To address this need, we developed a novel mouse ocular xenograft model. Green fluorescent protein (GFP) expressing human glioblastoma cells (between 500 and 10,000) were implanted into the subretinal space of immunodeficient mice (56 eyes). The resultant xenografts were imaged in vivo non-invasively with combined fluorescence scanning laser ophthalmoscopy (SLO) and volumetric optical coherence tomography (OCT) for a period up to several months. Most xenografts exhibited a latent phase followed by a stable or rapidly increasing volume, but about 1/3 underwent spontaneous remission. After prescribed growth, a population of tumors was treated with intravenously delivered doxorubicin-containing porphyrin and cholic acid-based nanoparticles ("nanodox"). Fluorescence resonance energy transfer (FRET) emission (doxorubicin → porphyrin) was used to localize nanodox in the xenografts, and 690 nm light exposure to activate it. Such photo-nanotherapy was highly effective in reducing tumor volume. Histopathology and flow cytometry revealed CD4 + and CD8 + immune cell infiltration of xenografts. Overall, the ocular model shows potential for examining the relationships between neoplastic growth, neovascularization and other features of the immune microenvironment, and for evaluating treatment response longitudinally in vivo.

2.
J Neuroinflammation ; 14(1): 121, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28645275

RESUMO

BACKGROUND: Retinal detachment (RD) can lead to proliferative vitreoretinopathy (PVR), a leading cause of intractable vision loss. PVR is associated with a cytokine storm involving common proinflammatory molecules like IL6, but little is known about the source and downstream signaling of IL6 and the consequences for the retina. Here, we investigated the early immune response and resultant cytokine signaling following RD in mice. METHODS: RD was induced in C57BL/6 J and IL6 knockout mice, and the resulting inflammatory response was examined using immunohistochemistry and flow cytometry. Cytokines and signaling proteins of vitreous and retinas were quantified by multiple cytokine arrays and Western blotting. To attempt to block IL6 signaling, a neutralizing antibody of IL6 receptor α (IL6Rα) or IL6 receptor ß (gp-130) was injected intravitreally immediately after RD. RESULTS: Within one day of RD, bone marrow-derived Cd11b + monocytes had extravasated from the vasculature and lined the vitreal surface of the retina, while the microglia, the resident macrophages of the retina, were relatively unperturbed. Cytokine arrays and Western blot analysis revealed that this sterile inflammation did not cause activation of IL6 signaling in the neurosensory retina, but rather only in the vitreous and aqueous humor. Monocyte infiltration was inhibited by blocking gp130, but not by IL6 knockout or IL6Rα blockade. CONCLUSIONS: Together, our results demonstrate that monocytes are the primary immune cell mediating the cytokine storm following RD, and that any resulting retinal damage is unlikely to be a direct result of retinal IL6 signaling, but rather gp130-mediated signaling in the monocytes themselves. These results suggest that RD should be treated immediately, and that gp130-directed therapies may prevent PVR and promote retinal healing.


Assuntos
Receptor gp130 de Citocina/metabolismo , Interleucina-6/metabolismo , Monócitos/metabolismo , Descolamento Retiniano/metabolismo , Transdução de Sinais/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Distribuição Aleatória , Descolamento Retiniano/patologia , Fatores de Tempo
3.
Hum Mol Genet ; 24(6): 1755-63, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25416279

RESUMO

Mutations in RPE65 or lecithin-retinol acyltransferase (LRAT) disrupt 11-cis-retinal synthesis and cause Leber congenital amaurosis (LCA), a severe hereditary blindness occurring in early childhood. The pathology is attributed to a combination of 11-cis-retinal deficiency and photoreceptor degeneration. The mistrafficking of cone membrane-associated proteins including cone opsins (M- and S-opsins), cone transducin (Gαt2), G-protein-coupled receptor kinase 1 (GRK1) and guanylate cyclase 1 (GC1) has been suggested to play a role in cone degeneration. However, their precise role in cone degeneration is unclear. Here we investigated the role of S-opsin (Opn1sw) in cone degeneration in Lrat(-) (/-), a murine model for LCA, by genetic ablation of S-opsin. We show that deletion of just one allele of S-opsin from Lrat(-) (/-) mice is sufficient to prevent the rapid cone degeneration for at least 1 month. Deletion of both alleles of S-opsin prevents cone degeneration for an extended period (at least 12 months). This genetic prevention is accompanied by a reduction of endoplasmic reticulum (ER) stress in Lrat(-) (/-) photoreceptors. Despite cone survival in Opn1sw(-/-)Lrat(-) (/-) mice, cone membrane-associated proteins (e.g. Gαt2, GRK1 and GC1) continue to have trafficking problems. Our results suggest that cone opsins are the 'culprit' linking 11-cis-retinal deficiency to cone degeneration in LCA. This result has important implications for the current gene therapy strategy that emphasizes the need for a combinatorial therapy to both improve vision and slow photoreceptor degeneration.


Assuntos
Deleção de Genes , Amaurose Congênita de Leber/patologia , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Degeneração Retiniana/patologia , Opsinas de Bastonetes/genética , Animais , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/genética , Amaurose Congênita de Leber/genética , Camundongos , Transporte Proteico , Degeneração Retiniana/genética , Degeneração Retiniana/prevenção & controle
4.
J Gen Physiol ; 135(3): 173-96, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20176852

RESUMO

Transport of proteins to and from cilia is crucial for normal cell function and survival, and interruption of transport has been implicated in degenerative and neoplastic diseases. It has been hypothesized that the ciliary axoneme and structures adjacent to and including the basal bodies of cilia impose selective barriers to the movement of proteins into and out of the cilium. To examine this hypothesis, using confocal and multiphoton microscopy we determined the mobility of the highly soluble photoactivatable green fluorescent protein (PAGFP) in the connecting cilium (CC) of live Xenopus retinal rod photoreceptors, and in the contiguous subcellular compartments bridged by the CC, the inner segment (IS) and the outer segment (OS). The estimated axial diffusion coefficients are D(CC) = 2.8 +/- 0.3, D(IS) = 5.2 +/- 0.6, and D(OS) = 0.079 +/- 0.009 microm(2) s(-1). The results establish that the CC does not pose a major barrier to protein diffusion within the rod cell. However, the results also reveal that axial diffusion in each of the rod's compartments is substantially retarded relative to aqueous solution: the axial diffusion of PAGFP was retarded approximately 18-, 32- and 1,000-fold in the IS, CC, and OS, respectively, with approximately 20-fold of the reduction in the OS attributable to tortuosity imposed by the lamellar disc membranes. Previous investigation of PAGFP diffusion in passed, spherical Chinese hamster ovary cells yielded D(CHO) = 20 microm(2) s(-1), and estimating cytoplasmic viscosity as D(aq)/D(CHO) = 4.5, the residual 3- to 10-fold reduction in PAGFP diffusion is ascribed to sub-optical resolution structures in the IS, CC, and OS compartments.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Cílio Conector dos Fotorreceptores/metabolismo , Segmento Interno das Células Fotorreceptoras da Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Algoritmos , Animais , Animais Geneticamente Modificados , Transporte Biológico , Difusão , Corantes Fluorescentes/metabolismo , Líquido Intracelular , Modelos Biológicos , Xenopus laevis
5.
N Engl J Med ; 358(21): 2240-8, 2008 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-18441370

RESUMO

Leber's congenital amaurosis (LCA) is a group of inherited blinding diseases with onset during childhood. One form of the disease, LCA2, is caused by mutations in the retinal pigment epithelium-specific 65-kDa protein gene (RPE65). We investigated the safety of subretinal delivery of a recombinant adeno-associated virus (AAV) carrying RPE65 complementary DNA (cDNA) (ClinicalTrials.gov number, NCT00516477 [ClinicalTrials.gov]). Three patients with LCA2 had an acceptable local and systemic adverse-event profile after delivery of AAV2.hRPE65v2. Each patient had a modest improvement in measures of retinal function on subjective tests of visual acuity. In one patient, an asymptomatic macular hole developed, and although the occurrence was considered to be an adverse event, the patient had some return of retinal function. Although the follow-up was very short and normal vision was not achieved, this study provides the basis for further gene therapy studies in patients with LCA.


Assuntos
Cegueira/terapia , Proteínas de Transporte/genética , Proteínas do Olho/genética , Terapia Genética , Vetores Genéticos , Degeneração Retiniana/terapia , Adulto , Cegueira/congênito , Cegueira/genética , Cegueira/patologia , DNA Complementar , Dependovirus/genética , Técnicas de Transferência de Genes , Humanos , Injeções , Mutação , Regiões Promotoras Genéticas , Reflexo Pupilar , Retina/patologia , Degeneração Retiniana/congênito , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Acuidade Visual , cis-trans-Isomerases
7.
Mol Ther ; 9(2): 182-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14759802

RESUMO

The congenital retinal blindness known as Leber congenital amaurosis (LCA) can be caused by mutations in the RPE65 gene. RPE65 plays a critical role in the visual cycle that produces the photosensitive pigment rhodopsin. Recent evidence from human studies of LCA indicates that earlier rather than later intervention may be more likely to restore vision. We determined the impact of in utero delivery of the human RPE65 cDNA to retinal pigment epithelium cells in a murine model of LCA, the Rpe65(-/-) mouse, using a serotype 2 adeno-associated virus packaged within an AAV1 capsid (AAV2/1). Delivery of AAV2/1-CMV-hRPE65 to fetuses (embryonic day 14) resulted in efficient transduction of retinal pigment epithelium, restoration of visual function, and measurable rhodopsin. The results demonstrate AAV-mediated correction of the deficit and suggest that in utero retinal gene delivery may be a useful approach for treating a variety of blinding congenital retinal diseases.


Assuntos
Cegueira/congênito , Cegueira/terapia , Modelos Animais de Doenças , Embrião de Mamíferos/metabolismo , Terapia Genética/métodos , Visão Ocular/fisiologia , Animais , Cegueira/genética , Cegueira/fisiopatologia , Proteínas de Transporte , Dependovirus/genética , Eletrorretinografia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/fisiologia , Embrião de Mamíferos/fisiopatologia , Proteínas do Olho , Feminino , Vetores Genéticos/genética , Camundongos , Camundongos Knockout , Proteínas/genética , Proteínas/metabolismo , Retina/embriologia , Retina/metabolismo , Retina/fisiologia , Útero , Visão Ocular/genética , cis-trans-Isomerases
8.
J Virol ; 77(14): 7957-63, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12829835

RESUMO

The development of fetal ocular gene transfer may be useful as a therapeutic tool for the prevention of retinal genetic disorders with congenital or early clinical manifestations. In this study we explored the neural progenitor transduction patterns of adeno-associated virus (AAV) vectors following delivery to the developing retina. Recombinant vectors with the same genome carrying the enhanced green fluorescent protein (EGFP) transgene packaged in capsids of differing serotypes (serotypes 1, 2, and 5, termed AAV2/1, AAV2/2, and AAV2/5, respectively) were created. Delivery of the AAV vectors during early retinal development resulted in efficient and stable transduction of retinal progenitors. Vector surface proteins and the developmental status of the retina profoundly affected viral tropism and transgene distribution. The procedure is not detrimental to retinal development and function and therefore provides a safe delivery vehicle for potential therapeutic applications and a means of assessing the mechanisms of retina development and disease.


Assuntos
Proteínas do Capsídeo/genética , Dependovirus/genética , Vetores Genéticos , Neurônios/fisiologia , Retina/embriologia , Transdução Genética , Animais , Diferenciação Celular , Dependovirus/classificação , Feminino , Técnicas de Transferência de Genes , Terapia Genética , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/virologia , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/embriologia , Gravidez , Retina/citologia , Retina/virologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Células-Tronco/virologia , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA