Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Ageing Res Rev ; 99: 102358, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38830546

RESUMEN

OBJECTIVE: The aim of this review article is to summarize the latest findings and current understanding of the origin of melanin in the retinal pigment epithelium (RPE), its function within the RPE, its role in the pathogenesis of age-related macular degeneration (AMD), its effect on retinal development, and its potential therapeutic benefit in the treatment of AMD. METHODS: A comprehensive search of peer-reviewed journals was conducted using various combinations of key terms such as "melanin," "retinal pigment epithelium" or "RPE," "age-related macular degeneration" or AMD," "lipofuscin," "oxidative stress," and "albinism." Databases searched include PubMed, Scopus, Science Direct, and Google Scholar. 147 papers published between the years of 1957 and 2023 were considered with an emphasis on recent findings. SUMMARY OF FINDINGS: AMD is thought to result from chronic oxidative stress within the RPE that results in cellular dysfunction, metabolic dysregulation, inflammation, and lipofuscin accumulation. Melanin functions as a photoscreener, free radical scavenger, and metal cation binding reservoir within the RPE. RPE melanin does not regenerate, and it undergoes degradation over time in response to chronic light exposure and oxidative stress. RPE melanin is important for retinal development and RPE function, and in the aging eye, melanin loss is associated with increased lipid peroxidation, inflammation, and the accumulation of toxic oxidized cellular products. Therefore, melanin-based treatments may serve to preserve RPE and retinal function in AMD. CONCLUSIONS: The pathogenesis of AMD is not fully understood, but RPE dysfunction and melanin loss in response to chronic oxidative stress and inflammation are thought to be primary drivers of the disease. Due to melanin's antioxidative effects, melanin-based nanotechnology represents a promising avenue for the treatment of AMD.

2.
Adv Sci (Weinh) ; : e2400230, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816934

RESUMEN

Exposure of the eyes to blue light can induce the overproduction of reactive oxygen species (ROS) in the retina and retinal pigment epithelium (RPE) cells, potentially leading to pathological damage of age-related macular degeneration (AMD). While the melanin in RPE cells absorbs blue light and prevents ROS accumulation, the loss and dysfunction of RPE melanin due to age-related changes may contribute to photooxidation toxicity. Herein, a novel approach utilizing a polydopamine-replenishing strategy via a single-dose intravitreal (IVT) injection is presented to protect retinal cells against blue light-induced phototoxicity. To investigate the effects of overexposure to blue light on retinal cells, a blue light exposure Nrf2-deficient mouse model is created, which is susceptible to light-induced retinal lesions. After blue light irradiation, retina degeneration and an overproduction of ROS are observed. The polydopamine-replenishing strategy demonstrated effectiveness in maintaining retinal structural integrity and preventing retina degeneration by reducing ROS production in retinal cells and limiting the phototoxicity of blue light exposure. These findings highlight the potential of polydopamine as a simple and effective replenishment for providing photoprotection against high-energy blue light exposure.

3.
Gene Ther ; 30(7-8): 628-640, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36935427

RESUMEN

Gene therapy for autosomal dominant retinitis pigmentosa (adRP) is challenged by the dominant inheritance of the mutant genes, which would seemingly require a combination of mutant suppression and wild-type replacement of the appropriate gene. We explore the possibility that delivery of a nanoparticle (NP)-mediated full-length mouse genomic rhodopsin (gRho) or human genomic rhodopsin (gRHO) locus can overcome the dominant negative effects of the mutant rhodopsin in the clinically relevant P23H+/--knock-in heterozygous mouse model. Our results demonstrate that mice in both gRho and gRHO NP-treated groups exhibit significant structural and functional recovery of the rod photoreceptors, which lasted for 3 months post-injection, indicating a promising reduction in photoreceptor degeneration. We performed miRNA transcriptome analysis using next generation sequencing and detected differentially expressed miRNAs as a first step towards identifying miRNAs that could potentially be used as rhodopsin gene expression enhancers or suppressors for sustained photoreceptor rescue. Our results indicate that delivering an intact genomic locus as a transgene has a greater chance of success compared to the use of the cDNA for treatment of this model of adRP, emphasizing the importance of gene augmentation using a gDNA that includes regulatory elements.


Asunto(s)
MicroARNs , Nanopartículas , Retinitis Pigmentosa , Ratones , Animales , Humanos , Rodopsina/genética , Rodopsina/química , Rodopsina/metabolismo , Modelos Animales de Enfermedad , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Genómica , MicroARNs/genética , Mutación
4.
ACS Nano ; 16(11): 19412-19422, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36269679

RESUMEN

Melanin is a natural pigment that is widely distributed in many parts of the human body, such as the skin and retinal pigment epithelium (RPE) in eyes. In contrast to skin melanin, which is being constantly synthesized by the epidermal melanocytes, melanin in the RPE does not regenerate. Melanin is known to function as a potential radical scavenger and photoprotective agent. However, the protective effects of melanin against oxidative stress decline with increasing age. This phenomenon has been correlated with the pathogenesis of age-related macular degeneration (AMD). To increase the potential antioxidant and photoprotective characteristics of melanin, we designed a therapeutic strategy for replenishment of melanin using PEGylated synthetic melanin-like nanoparticles (MNPs) in the RPE for the treatment of AMD. We performed experiments using AMD-like cellular and mouse models and demonstrated that MNPs are biocompatible and selectively target reactive oxygen species (ROS) with powerful antioxidant properties. MNPs can traffic and accumulate in the RPE and are exclusively located in cytosol, but not the nucleus and mitochondria of the cells, for at least 3 months after a single-dose intravitreal injection. Our findings demonstrate that MNPs are able to substitute for natural melanin in the RPE and suggest the potential efficacy of MNPs as a natural radical scavenger against oxidative stress in ROS-related diseases, such as AMD.


Asunto(s)
Degeneración Macular , Nanopartículas , Ratones , Animales , Humanos , Epitelio Pigmentado de la Retina/patología , Especies Reactivas de Oxígeno/farmacología , Melaninas , Antioxidantes/farmacología , Degeneración Macular/tratamiento farmacológico , Estrés Oxidativo
5.
Curr Gene Ther ; 22(2): 89-103, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33874871

RESUMEN

Gene therapy has made significant development since the commencement of the first clinical trials a few decades ago and has remained a dynamic area of research regardless of obstacles such as immune response and insertional mutagenesis. Progression in various technologies like next-generation sequencing (NGS) and nanotechnology has established the importance of non-- coding segments of a genome, thereby taking gene therapy to the next level. In this review, we have summarized the importance of non-coding elements, highlighting the advantages of using full- length genomic DNA loci (gDNA) compared to complementary DNA (cDNA) or minigene, currently used in gene therapy. The focus of this review is to provide an overview of the advances and the future of potential use of gDNA loci in gene therapy, expanding the therapeutic repertoire in molecular medicine.


Asunto(s)
ADN , Genoma , ADN/genética , ADN Complementario , Terapia Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ADN
6.
Nature ; 581(7806): 83-88, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32376950

RESUMEN

Photoreceptor loss is the final common endpoint in most retinopathies that lead to irreversible blindness, and there are no effective treatments to restore vision1,2. Chemical reprogramming of fibroblasts offers an opportunity to reverse vision loss; however, the generation of sensory neuronal subtypes such as photoreceptors remains a challenge. Here we report that the administration of a set of five small molecules can chemically induce the transformation of fibroblasts into rod photoreceptor-like cells. The transplantation of these chemically induced photoreceptor-like cells (CiPCs) into the subretinal space of rod degeneration mice (homozygous for rd1, also known as Pde6b) leads to partial restoration of the pupil reflex and visual function. We show that mitonuclear communication is a key determining factor for the reprogramming of fibroblasts into CiPCs. Specifically, treatment with these five compounds leads to the translocation of AXIN2 to the mitochondria, which results in the production of reactive oxygen species, the activation of NF-κB and the upregulation of Ascl1. We anticipate that CiPCs could have therapeutic potential for restoring vision.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Degeneración Retiniana/terapia , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/trasplante , Visión Ocular/efectos de los fármacos , Animales , Proteína Axina/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Visión Ocular/fisiología
7.
Nat Cancer ; 1(10): 990-997, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33898999

RESUMEN

Retinoblastoma (RB) is a pediatric retinal tumor that overexpresses the ganglioside GD2. Although it is treatable in patients with early diagnosis, patients may lose one or two eyes. We generated GD2-specific chimeric antigen receptor T lymphocytes (GD2.CAR-Ts) and locally delivered them to mice with an in-situ grafting RB. When used in combination with the local release of interleukin (IL)-15 and an injectable hydrogel, we showed that GD2.CAR-Ts successfully eliminate RB tumor cells without impairment of the mouse vision.


Asunto(s)
Neoplasias de la Retina , Retinoblastoma , Animales , Gangliósidos , Humanos , Hidrogeles , Ratones , Neoplasias de la Retina/terapia , Retinoblastoma/terapia , Linfocitos T
8.
Mol Ther ; 28(2): 523-535, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31879189

RESUMEN

The use of gene therapy may allow replacement of the defective gene. Minigenes, such as cDNAs, are often used. However, these may not express normal physiological genetic profiles due to lack of crucial endogenous regulatory elements. We constructed DNA nanoparticles (NPs) that contain either the mouse or human full-length rhodopsin genomic locus, including endogenous promoters, all introns, and flanking regulatory sequences of the 15-16 kb genomic rhodopsin DNA inserts. We transduced the NPs into primary retinal cell cultures from the rhodopsin knockout (RKO) mouse in vitro and into the RKO mouse in vivo and compared the effects on different functions to plasmid cDNA NP counterparts that were driven by ubiquitous promoters. Our results demonstrate that genomic DNA vectors resulted in long-term high levels of physiological transgene expression over a period of 5 months. In contrast, the cDNA counterparts exhibited low levels of expression with sensitivity to the endoplasmic reticulum (ER) stress mechanism using the same transgene copy number both in vitro and in vivo. This study demonstrates for the first time the transducing of the rhodopsin genomic locus using compacted DNA NPs.


Asunto(s)
ADN/administración & dosificación , Expresión Génica , Terapia Genética , Nanopartículas , Degeneración Retiniana/genética , Rodopsina/genética , Animales , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Técnicas de Transferencia de Gen , Humanos , Ratones , Ratones Noqueados , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneración Retiniana/patología , Degeneración Retiniana/terapia , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/terapia , Transgenes
9.
J Control Release ; 315: 40-54, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31669212

RESUMEN

Dry eye (DE) disease is an uprising health epidemic that directly affects the surface of the eye. We developed a water soluble cerium oxide loaded glycol chitosan nanoparticle as a new type of eye drop, namely GCCNP (glycol chitosan cerium oxide nanoparticles). GCCNP is capable of scavenging cellular reactive oxygen species (ROS) for the treatment of DE disease. The antioxidative effects of GCCNP were assessed in mice primary corneal and conjunctival cells in vitro and in a DE murine model in vivo. GCCNP's effect on the DE models was assessed via histological evaluations, migration assays, cell viability assays, cellular uptake analyses, intracellular ROS scavenging assays, wound healing assays, mitochondrial membrane potential readings, corneal fluorescein staining, tear volume concentrations, tear film break up time analyses, and lastly, analytical/spectroscopic analyses of GCCNP eye drop formulations. Spectroscopic analysis showed that cerium oxide was entrapped into the glycol chitosan (GC). The solubility of cerium in GC (GCCNP) increased to 709.854±24.3µg/ml compared to its original solubility in cerium oxide, which was measured as 0.020±0.002µg/ml. GCCNP had no cytotoxic effect and showed improvements on dry eye disease models by stabilizing the tear film, scavenging ROS, up-regulating SOD, promoting and maintaining corneal and conjunctival cell growth and integrity. We provided convincing evidence that GCCNP is an effective treatment for DE and may represent a potential new class of drug for DE disease.


Asunto(s)
Cerio/administración & dosificación , Quitosano/química , Síndromes de Ojo Seco/tratamiento farmacológico , Nanopartículas , Animales , Antioxidantes/administración & dosificación , Antioxidantes/química , Antioxidantes/farmacología , Cerio/química , Cerio/farmacología , Conjuntiva/citología , Conjuntiva/efectos de los fármacos , Córnea/citología , Córnea/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Solubilidad , Superóxido Dismutasa/metabolismo , Lágrimas/efectos de los fármacos
10.
Sci Rep ; 9(1): 14573, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601909

RESUMEN

Elevated oxidative stress and associated reactive oxygen species (ROS) accumulation are hallmarks in the induction and progression of age-related macular degeneration (AMD). By exposing nuclear factor erythroid 2-related factor (Nrf2) knockout (Nrf2-/-) mice to mild white light, we were able to generate a new dry-AMD like murine model to the study. This animal model developed phenotypes of photoreceptor degeneration, retinal function impairment, ROS accumulation, and inflammation reaction in a relatively shorter time. In the treatment of this animal model we utilized an antioxidative and water soluble nanoparticle known as glycol chitosan coated cerium oxide nanoparticles (GCCNP). The delivery of GCCNP protected retina against progressive retinal oxidative damage. Further combination of GCCNP with alginate-gelatin based injectable hydrogel provided synergistic antioxidant effects and achieved a more rapid recovery of the retinal pigment epithelium and photoreceptor cells. This combined treatment technique has the potential to translate into a clinical intervention for the treatment of AMD.


Asunto(s)
Cerio/uso terapéutico , Hidrogeles/química , Luz , Degeneración Macular/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/genética , Animales , Antioxidantes/metabolismo , Quitosano/química , Electrorretinografía , Radicales Libres , Regulación de la Expresión Génica , Atrofia Geográfica/tratamiento farmacológico , Glicoles/química , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/química , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Retina/fisiopatología
11.
J Biomed Mater Res A ; 106(11): 2795-2804, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29752862

RESUMEN

The major purpose of this article is to evaluate oligochitosan coated cerium oxide nanoparticles (OCCNPs) alginate laden injectable hydrogels and their potential treatment for age-related macular degeneration (AMD). The water soluble OCCNPs were loaded within injectable hydrogels as antioxidative agents. The release of OCCNPs from hydrogel, radical scavenging properties, and biocompatibility were evaluated and calculated in vitro. The effects of OCCNP laden hydrogel downregulating expression of angiogenic proteins and proinflammatory cytokines were quantified in human retinal pigment epithlium-19 (ARPE-19) and umbilical endothelium cell lines. The hydrogels behaved with moderate swelling and controllable degradation. The laden OCCNPs were released in a controlled manner in vitro during two months of testing. The OCCNP loaded hydrogels exhibited robust antioxidative properties in oxygen radical absorbance capacity tests and reduced apoptosis in H2 O2 -induced ARPE-19 cells. Furthermore, OCCNP loaded injectable hydrogels are biocompatible and suppressed the ipopolysaccharides-induced inflammation response in ARPE-19 cells, and inhibited expression of vascular endothelium growth factor in human ARPE-19 and umbilical endothelium cell lines. The alginate-gelatin injectable hydrogel loaded OCCNPs are biocompatible and have high potential in protecting cells from apoptosis, angiogenesis, and production of proinflammatory cytokines in AMD cellular models. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2795-2804, 2018.


Asunto(s)
Alginatos/química , Inhibidores de la Angiogénesis/administración & dosificación , Antioxidantes/administración & dosificación , Cerio/administración & dosificación , Portadores de Fármacos/química , Hidrogeles/química , Degeneración Macular/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Antioxidantes/farmacología , Materiales Biocompatibles/química , Línea Celular , Cerio/farmacología , Quitosano/química , Sistemas de Liberación de Medicamentos , Gelatina/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Fisiológica/efectos de los fármacos
12.
Int J Nanomedicine ; 13: 1361-1379, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29563793

RESUMEN

INTRODUCTION: DNA nanoparticles (NPs) comprising polylysine conjugated to polyethylene glycol efficiently target murine photoreceptors and the retinal pigment epithelium (RPE) and lead to long-term phenotypic improvement in models of retinal degeneration. Advancing this technology requires testing in a large animal model, particularly with regard to safety. So, herein we evaluate NPs in non-human primates (baboon). METHODS AND RESULTS: NPs with plasmids carrying GFP and a ubiquitous, RPE-specific, or photoreceptor-specific promoter were delivered by either subretinal or intravitreal injection. We detected GFP message and protein in the retina/RPE from eyes dosed with NPs carrying ubiquitously expressed and RPE-specific vectors, and GFP message in eyes injected with NPs carrying photoreceptor-specific vectors. Importantly, we observed NP DNA in the retina/RPE following intravitreal injection, indicating the inner limiting membrane does not prevent NP diffusion into the outer retina. We did not observe any adverse events in any baboon, and there were no NP-associated changes in retinal function. Furthermore, no systemic or local inflammatory reaction to the vectors/injections was observed, and no NP DNA was found outside the eye. CONCLUSION: Taken together with the well-established rodent safety and efficacy data, these findings suggest that DNA NPs may be a safe and potentially clinically viable nonviral ocular therapy platform for retinal diseases.


Asunto(s)
ADN/química , Ojo/efectos de los fármacos , Nanopartículas/toxicidad , Animales , Encéfalo/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Inyecciones Intravítreas , Ratones , Nanopartículas/química , Plásmidos/metabolismo , Primates , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo
13.
Invest Ophthalmol Vis Sci ; 59(2): 746-756, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29392320

RESUMEN

Purpose: Our goal was to generate and characterize a new mouse model in which only angiogenesis- and glaucoma-relevant tissues would be naturally fluorescent. The Matrix Gla (MGP) gene is highly expressed in vascular smooth muscle cells (VSMC) and trabecular meshwork (TM). We sought to direct our Mgp-Cre.KI mouse recombinase to VSMC/TM cells to produce their longitudinal fluorescent profiles. Methods: Homozygous Mgp-Cre.KI mice were crossed with Ai9 homozygous reporter mice harboring a loxP-flanked STOP cassette preventing transcription of a DsRed fluorescent protein (tdTomato). The F1 double-heterozygous (Mgp-tdTomato) was examined by direct fluorescence, whole mount, histology, and fundus photography. Custom-made filters had 554/23 emission and 609/54 exciter nanometer wavelengths. Proof of concept of the model's usefulness was conducted by inducing guided imaging laser burns. Evaluation of a vessel's leakage and proliferation was followed by noninvasive angiography. Results: The Mgp-tdTomato mouse was viable, fertile, with normal IOP and ERG. Its phenotype exhibited red paws and snout (cartilage expression), which precluded genotyping. A fluorescent red ring was seen at the limbus and confirmed to be TM expression by histology. The entire retinal vasculature was red fluorescent (VSMC) and directly visualized by fundus photography. Laser burns on the Mgp-tdTomato allowed separation of leakiness and neovascularization evaluation parameters. Conclusions: The availability of a transgenic mouse naturally fluorescent in glaucoma-relevant tissues and retinal vasculature brings the unique opportunity to study a wide spectrum of single and combined glaucomatous conditions in vivo. Moreover, the Mgp-tdTomato mouse provides a new tool to study mechanisms and therapeutics of retinal angiogenesis longitudinally.


Asunto(s)
Proteínas de Unión al Calcio/genética , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Regulación de la Expresión Génica/fisiología , Glaucoma/genética , Proteínas Luminiscentes/genética , Neovascularización Retiniana/genética , Malla Trabecular/metabolismo , Animales , Neovascularización Coroidal/genética , Cruzamientos Genéticos , Electrorretinografía , Femenino , Colorantes Fluorescentes , Integrasas , Presión Intraocular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Retina/fisiología , Neovascularización Retiniana/fisiopatología , Proteína Gla de la Matriz
14.
Adv Funct Mater ; 28(52)2018 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-32699541

RESUMEN

Development of a single combinatorial nano-platform technology to target cancer cells has been an unprecedented reality in boosting synergistic anti-tumor activities and in reducing off-target effects. We have designed a novel anti-tumor delivery system using a chemotherapy drug and a tumor target molecule covalently linked to cerium oxide nanoparticles (nanoceria). Nanoceria have a unique redox activity in that they possess antioxidant activity at physiological pH but have an intrinsic oxidase activity at acidic pH. Our system is integrated with (1) extracellular pH responsive functionality, (2) tumor cell targetable (CXC chemokine receptor 4, CXCR4 receptor specific) antagonist, (3) reactive oxygen species (ROS) inducible nanoceria, and (4) chemotherapeutic doxorubicin (DOX). These combinatorial nanoparticles (AMD-GCCNPs-DOX) are not only sensitive to the extracellular acidic pH conditions and targeted tumor cells but can also instantaneously induce ROS and release DOX intracellularly to enhance the chemotherapeutic activity in retinoblastoma cells (WERI-Rb-1 and Y79) and in xenograft (Y79/GFP-luc grafted) and genetic p107s (Rb Lox/lox , p107 +/- , p130 -/- ) orthotopic mice models. Together we introduce a lucidly engineered combinatorial nano-construct that offers a viable and simple strategy for delivering a cocktail of therapeutics into tumor cells under acidosis, exhibiting a promising new future for clinical therapeutic opportunities.

15.
Biomaterials ; 157: 26-39, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29232624

RESUMEN

Retinitis pigmentosa (RP) is a group of inherited retinal degenerative conditions and a leading cause of irreversible blindness. 25%-30% of RP cases are caused by inherited autosomal dominant (ad) mutations in the rhodopsin (Rho) protein of the retina, which impose a barrier for developing therapeutic treatments for this genetically heterogeneous disorder, as simple gene replacement is not sufficient to overcome dominant disease alleles. Previously, we have explored using the genomic short-form of Rho (sgRho) for gene augmentation therapy of RP in a Rho knockout mouse model. We have shown improved gene expression and fewer epigenetic modifications compared with the use of a Rho cDNA expression construct. In the current study, we altered our strategy by delivering a codon-optimized genomic form of Rho (co-sgRho) (for gene replacement) in combination with an RNAi-based inactivation of endogenous Rho alleles (gene suppression of both mutant Rho alleles, but mismatched with the co-sgRho) into a homozygous RhoP23H/P23H knock-in (KI) RP mouse model, which has a severe phenotype of adRP. In addition, we have conjugated a cell penetrating TAT peptide sequence to our previously established CK30PEG10 diblock co-polymer. The DNAs were compacted with CK30PEG10-TAT diblock co-polymer to form DNA nanoparticles (NPs). These NPs were injected into the sub-retinal space of the KI mouse eyes. As a proof of concept, we demonstrated the efficiency of this strategy in the partial improvement of visual function in the RhoP23H/P23H KI mouse model.


Asunto(s)
ADN/administración & dosificación , Modelos Animales de Enfermedad , Terapia Genética/métodos , Nanopartículas/administración & dosificación , Retinitis Pigmentosa/terapia , Rodopsina/fisiología , Animales , ADN/química , Técnicas de Sustitución del Gen/métodos , Genes Dominantes , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Degeneración Retiniana , Retinitis Pigmentosa/genética
16.
J Control Release ; 268: 212-224, 2017 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-29061512

RESUMEN

The demand for effective eye therapies is driving the development of injectable hydrogels as new medical devices for controlled delivery and filling purposes. This article introduces the properties of injectable hydrogels and summarizes their versatile application in the treatment of ophthalmic diseases, including age-related macular degeneration, cataracts, diabetic retinopathy, glaucoma, and intraocular cancers. A number of injectable hydrogels are approved by FDA as surgery sealants, tissue adhesives, and are now being investigated as a vitreous humor substitute. Research on hydrogels for drug, factor, nanoparticle, and stem cell delivery is still under pre-clinical investigation or in clinical trials. Although substantial progress has been achieved using injectable hydrogels, some challenging issues must still be overcome before they can be effectively used in medical practice.


Asunto(s)
Sistemas de Liberación de Medicamentos , Oftalmopatías/tratamiento farmacológico , Hidrogeles/administración & dosificación , Animales , Humanos , Inyecciones
17.
ACS Nano ; 11(5): 4669-4685, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28463509

RESUMEN

Age-related macular degeneration (AMD) is the foremost cause of irreversible blindness in people over the age of 65 especially in developing countries. Therefore, an exploration of effective and alternative therapeutic interventions is an unmet medical need. It has been established that oxidative stress plays a key role in the pathogenesis of AMD, and hence, neutralizing oxidative stress is an effective therapeutic strategy for treatment of this serious disorder. Owing to autoregenerative properties, nanoceria has been widely used as a nonenzymatic antioxidant in the treatment of oxidative stress related disorders. Yet, its potential clinical implementation has been greatly hampered by its poor water solubility and lack of reliable tracking methodologies/processes and hence poor absorption, distribution, and targeted delivery. The water solubility and surface engineering of a drug with biocompatible motifs are fundamental to pharmaceutical products and precision medicine. Here, we report an engineered water-soluble, biocompatible, trackable nanoceria with enriched antioxidant activity to scavenge intracellular reactive oxygen species (ROS). Experimental studies with in vitro and in vivo models demonstrated that this antioxidant is autoregenerative and more active in inhibiting laser-induced choroidal neovascularization by decreasing ROS-induced pro-angiogenic vascular endothelial growth factor (VEGF) expression, cumulative oxidative damage, and recruitment of endothelial precursor cells without exhibiting any toxicity. This advanced formulation may offer a superior therapeutic effect to deal with oxidative stress induced pathogeneses, such as AMD.


Asunto(s)
Cerio/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/farmacología , Cerio/química , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/prevención & control , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Degeneración Macular/fisiopatología , Degeneración Macular/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
J Control Release ; 236: 31-7, 2016 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-27297781

RESUMEN

We recently reported that the Ins2(Akita) mouse is a good model for late-onset diabetic retinopathy. Here, we investigated the effect of miR200-b, a potential anti-angiogenic factor, on VEGF receptor 2 (VEGFR-2) expression and to determine the underlying angiogenic response in mouse endothelial cells, and in retinas from aged Ins2(Akita) mice. MiR200-b and its native flanking sequences were amplified and cloned into a pCAG-eGFP vector directed by the ubiquitous CAG promoter (namely pCAG-miR200-b-IRES-eGFP). The plasmid was compacted by CK30PEG10K into DNA nanoparticles (NPs) for in vivo delivery. Murine endothelial cell line, SVEC4-10, was first transfected with the plasmid. The mRNA levels of VEGF and VEGFR-2 were quantified by qRT-PCR and showed significant reduction in message expression compared with lipofectamine-transfected cells. Transfection of miR200-b suppressed the migration of SVEC4-10 cells. There was a significant inverse correlation between the level of expression of miR200-b and VEGFR-2. Intravitreal injection of miR200-b DNA NPs significantly reduced protein levels of VEGFR-2 as revealed by western blot and markedly suppressed angiogenesis as evaluated by fundus imaging in aged Ins2(Akita) mice even after 3months of post-injection. These findings suggest that NP-mediated miR200-b delivery has negatively regulated VEGFR-2 expression in vivo.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , MicroARNs/administración & dosificación , Nanopartículas/química , Neovascularización Fisiológica , Animales , Línea Celular , Movimiento Celular , Diabetes Mellitus Experimental/complicaciones , Retinopatía Diabética/etiología , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Inyecciones Intravítreas , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Péptidos/química , Polietilenglicoles/química , Transfección/métodos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
19.
FASEB J ; 30(3): 1076-86, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26564956

RESUMEN

Previously, we compared the efficacy of nanoparticle (NP)-mediated intron-containing rhodopsin (sgRho) vs. intronless cDNA in ameliorating retinal disease phenotypes in a rhodopsin knockout (RKO) mouse model of retinitis pigmentosa. We showed that NP-mediated sgRho delivery achieved long-term expression and phenotypic improvement in RKO mice, but not NP housing cDNA. However, the protein level of the NP-sgRho construct was only 5-10% of wild-type at 8 mo postinjection. To have a better understanding of the reduced levels of long-term expression of the vectors, in the present study, we evaluated the epigenetic changes of subretinal delivering NP-cDNA vs. NP-sgRho in the RKO mouse eyes. Following the administration, DNA methylation and histone status of specific regions (bacteria plasmid backbone, promoter, rhodopsin gene, and scaffold/matrix attachment region) of the vectors were evaluated at various time points. We documented that epigenetic transgene silencing occurred in vector-mediated gene transfer, which were caused by the plasmid backbone and the cDNA of the transgene, but not the intron-containing transgene. No toxicity or inflammation was found in the treated eyes. Our results suggest that cDNA of the rhodopsin transgene and bacteria backbone interfered with the host defense mechanism of DNA methylation-mediated transgene silencing through heterochromatin-associated modifications.


Asunto(s)
ADN Complementario/genética , ADN/genética , Silenciador del Gen/fisiología , Intrones/genética , Nanopartículas/administración & dosificación , Rodopsina/genética , Transgenes/genética , Animales , Metilación de ADN/genética , Modelos Animales de Enfermedad , Expresión Génica/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Retinitis Pigmentosa/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-26109528

RESUMEN

Ophthalmic gene therapy is an intellectual and intentional manipulation of desired gene expression into the specific cells of an eye for the treatment of ophthalmic (ocular) genetic dystrophies and pathological conditions. Exogenous nucleic acids such as DNA, small interfering RNA, micro RNA, and so on, are used for the purpose of managing expression of the desired therapeutic proteins in ocular tissues. The delivery of unprotected nucleic acids into the cells is limited because of exogenous and endogenous degradation modalities. Nanotechnology, a promising and sophisticated cutting edge tool, works as a protective shelter for these therapeutic nucleic acids. They can be safely delivered to the required cells in order to modulate anticipated protein expression. To this end, nanotechnology is seen as a potential and promising strategy in the field of ocular gene delivery. This review focused on current nanotechnology modalities and other promising nonviral strategies being used to deliver therapeutic genes in order to treat various devastating ocular diseases.


Asunto(s)
Administración Oftálmica , Terapia Genética , Nanomedicina , Nanopartículas , Nanopartículas/administración & dosificación , Nanopartículas/química , Nanopartículas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...