Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Exp Eye Res ; 175: 20-31, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29864441

RESUMEN

There is currently no efficient method available for the delivery of full length functional proteins into the cytoplasm of retinal cells in vivo. Historically, the most successful approach for the treatment of retinal diseases has been intravitreal injection of antibodies or recombinant proteins, but this approach is not yet utilized for the delivery of proteins that require intracellular access for a therapeutic effect. Here we describe a platform for the delivery of functional proteins into ganglion cells, photoreceptors and retinal pigment epithelium via intravitreal injection. A nucleolin binding aptamer, AS1411, was biotinylated and complexed with traptavidin and utilized as a platform for the delivery of GFP or X-linked inhibitor of apoptosis (XIAP) proteins by intravitreal injection in BALB/c mice. Retinal sections were analyzed for uptake of proteins in the retina. Apoptosis was induced by intravitreal injection of N-methyl-D-aspartate (NMDA). Retinas were harvested for analysis of TUNEL and caspase 3/7 activity. Intravitreal injection of AS1411-directed GFP or XIAP complexes enabled delivery of these proteins into ganglion cells, photoreceptors and retinal pigment epithelium in vivo. AS1411-XIAP complexes conferred significant protection to cells in the outer and inner nuclear layers following NMDA induced apoptosis. A concomitant decrease in activity of Caspase 3/7 was observed in eyes injected with the AS1411-XIAP complex. In conclusion, AS1411 can be used as a platform for the delivery of therapeutic proteins into retinal cells. This approach can potentially be utilized to introduce a large variety of therapeutically relevant proteins that are previously well characterized to maintain the structural integrity and function of retina, thus, preventing vision loss due to ocular trauma or inherited retinal degeneration.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/administración & dosificación , Sistemas de Liberación de Medicamentos , Oligodesoxirribonucleótidos/administración & dosificación , Retina/efectos de los fármacos , Degeneración Retiniana/prevención & control , Proteína Inhibidora de la Apoptosis Ligada a X/administración & dosificación , Animales , Aptámeros de Nucleótidos/administración & dosificación , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Electroforesis en Gel de Poliacrilamida , Agonistas de Aminoácidos Excitadores/toxicidad , G-Cuádruplex , Proteínas Fluorescentes Verdes/administración & dosificación , Etiquetado Corte-Fin in Situ , Inyecciones Intravítreas , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , N-Metilaspartato/toxicidad , Degeneración Retiniana/patología
2.
Mol Ther ; 26(6): 1568-1580, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29678656

RESUMEN

Uveitis is an inflammatory disorder of the eye responsible for approximately 10%-15% of blindness in the US. In this study, we examined the role of the complement membrane attack complex (MAC) and the NLRP3 inflammasome in the pathogenesis of experimental autoimmune uveitis (EAU) in normal and C9-/- mice that are incapable of assembling the MAC. We discovered that the MAC and the NLRP3 inflammasome and associated production of IL-1ß are elevated in EAU mice and that MAC may be involved in regulation of Th1 and Th17 cell differentiation. In contrast, MAC and the NLRP3 inflammasome were not elevated in C9-/- mice. However, EAU-associated pathophysiology including retinal structure and function were not rescued in C9-/- mice. Unexpectedly, AAV-mediated delivery of sCD59, an inhibitor of C9 incorporation into the MAC, successfully attenuated activation of the NLRP3 inflammasome and EAU pathology as well as MAC. Our studies provide an improved understanding of the role of the MAC and the NLRP3 inflammasome in EAU as well as suggest a novel approach for the treatment of uveitis.


Asunto(s)
Antígenos CD59/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Uveítis/metabolismo , Animales , Antígenos CD59/genética , Activación de Complemento/genética , Activación de Complemento/fisiología , Inflamasomas/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Uveítis/genética
3.
Mol Ther Nucleic Acids ; 8: 77-89, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28918058

RESUMEN

Non-viral gene delivery systems are being developed to address limitations of viral gene delivery. Many of these non-viral systems are modeled on the properties of viruses including cell surface binding, endocytosis, endosomal escape, and nuclear targeting. Most non-viral gene transfer systems exhibit little correlation between in vitro and in vivo efficiency, hampering a systematic approach to their development. Previously, we have described a 3.5 kDa peptide (peptide for ocular delivery [POD]) that targets cell surface sialic acid. When functionalized with polyethylene glycol (PEG) via a sulfhydryl group on the N-terminal cysteine of POD, PEG-POD could compact plasmid DNA, forming 120- to 180-nm homogeneous nanoparticles. PEG-POD enabled modest gene transfer and rescue of retinal degeneration in vivo. Systematic investigation of different stages of gene transfer by PEG-POD nanoparticles was hampered by their inability to deliver genes in vitro. Herein, we describe functionalization of POD with PEG using a reducible orthopyridyl disulfide bond. These reducible nanoparticles enabled gene transfer in vitro while retaining their in vivo gene transfer properties. These reducible PEG-POD nanoparticles were utilized to deliver human FLT1 to the retina in vivo, achieving a 50% reduction in choroidal neovascularization in a murine model of age-related macular degeneration.

4.
Exp Eye Res ; 145: 380-392, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26923800

RESUMEN

There is currently no available method to efficiently deliver proteins across the plasma membrane of photoreceptor or retinal pigment epithelium (RPE) cells in vivo. Thus, current clinical application of recombinant proteins in ophthalmology is limited to the use of proteins that perform their biological function extracellularly. The ability to traverse biological membranes would enable the mobilization of a significantly larger number of proteins with previously well characterized properties. Nucleolin is abundantly present on the surface of rapidly dividing cells including cancer cells. Surprisingly, nucleolin is also present on the surface of photoreceptor cell bodies. Here we investigated whether nucleolin can be utilized as a gateway for the delivery of proteins into retinal cells following intravitreal injection. AS1411 is a G-quartet aptamer capable of targeting nucleolin. Subsequent to intravitreal injection, fluorescently labeled AS1411 localized to various retinal cell types including the photoreceptors and RPE. AS1411 linked to streptavidin (a ∼50 kDa protein) via a biotin bridge enabled the uptake of Streptavidin into photoreceptors and RPE. AS1411-Streptavidin conjugate applied topically to the cornea allowed for uptake of the conjugate into the nucleus and cytoplasm of corneal endothelial cells. Clinical relevance of AS1411 as a delivery vehicle was strongly indicated by demonstration of the presence of cell surface nucleolin on the photoreceptors, inner neurons and ganglion cells of human retina. These data support exploration of AS1411 as a means of delivering therapeutic proteins to diseased retina.


Asunto(s)
Aptámeros de Nucleótidos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Oligonucleótidos/administración & dosificación , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Recombinantes/administración & dosificación , Epitelio Pigmentado de la Retina/metabolismo , Análisis de Varianza , Animales , Humanos , Inmunohistoquímica , Inyecciones Intravítreas , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/metabolismo , Nucleolina
5.
Exp Eye Res ; 140: 171-178, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26368850

RESUMEN

Choroidal neovascularization (CNV) associated with the 'wet' form of age related macular degeneration (AMD) is one of the most common causes of central vision loss among the elderly. The 'wet' form of AMD is currently treated by intravitreal delivery of anti-VEGF agents. However, intravitreal injections are associated with complications and long-term inhibition of VEGF leads to macular atrophy. Thus, there is currently an unmet need for the development of therapies for CNV that target molecules other than VEGF. Here, we describe nucleolin as a novel target for the 'wet' form of AMD. Nucleolin was found on the surface of endothelial cells that migrate from the choroid into the subretinal space in the laser-induced model of 'wet' AMD. AS1411 is a previously described G-quartet oligonucleotide that has been shown to bind nucleolin. We found that AS1411 inhibited the formation of tubes by human umbilical vein endothelial cells (HUVECs) by approximately 27.4% in vitro. AS1411 co-localized with the site of laser induced CNV in vivo. Intravitreally injected AS1411 inhibited laser-induced CNV by 37.6% and attenuated infiltration of macrophages by 40.3%. Finally, topical application of AS1411 led to a 43.4% reduction in CNV. Our observations have potential implications for the development of therapies for CNV and specifically for the 'wet' form of AMD.


Asunto(s)
Aptámeros de Nucleótidos/administración & dosificación , Neovascularización Coroidal/prevención & control , Modelos Animales de Enfermedad , Oligodesoxirribonucleótidos/administración & dosificación , Fosfoproteínas/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Degeneración Macular Húmeda/prevención & control , Administración Tópica , Animales , Antígenos de Diferenciación/metabolismo , Movimiento Celular/efectos de los fármacos , Neovascularización Coroidal/metabolismo , Endotelio Vascular/efectos de los fármacos , Glicoesfingolípidos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inyecciones Intravítreas , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Microvasos/efectos de los fármacos , Neovascularización Patológica/prevención & control , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Degeneración Macular Húmeda/metabolismo , Nucleolina
6.
J Gene Med ; 17(10-12): 229-43, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26369397

RESUMEN

BACKGROUND: Age-related macular degeneration (AMD) is the most common cause of blindness in the elderly, with no therapy available for 90% of patients. Recent genetic evidence implicates activation of complement in the pathogenesis of AMD. We have recently discovered that adenovirus (Ad)-mediated expression of complement component C3 (AdCMVC3) in the murine retina recapitulates many of the pathological features found in human AMD. In the present study, utilizing a gene therapy approach, we examine whether Ad-mediated expression of complement Factor H (AdCAGfH) attenuates AdCMVC3-mediated retinal pathology. METHODS: AdCMVC3 was co-injected with either AdCAGfH or a negative control virus expressing green fluorescent protein (AdCMVGFP) into the subretinal space of adult mice. The resulting retinal pathology was analyzed by histology and immunocytochemistry and retinal function was quantified by electroretinography. RESULTS: Morphological and functional analyses indicated that AdCMVC3-mediated retinal pathology could be attenuated by AdCAGfH. Specifically, endothelial cell proliferation was reduced by 91% and atrophy of retinal pigment epithelium (RPE) could be attenuated by 69%. AdCAGfH injected eyes exhibited 90-150% greater A-wave and 120-180% greater B-wave amplitudes relative to control eyes. Immunocytochemical analysis of rhodopsin and RPE65 was consistent with the rescue of photoreceptors and RPE in AdCAGfH injected eyes. CONCLUSIONS: C3-induced pathology in murine retina can be attenuated by Ad-mediated expression of Factor H. Expression of Factor H is worthy of further study as a potential gene therapy for AMD.


Asunto(s)
Complemento C3/metabolismo , Factor H de Complemento/uso terapéutico , Adenoviridae , Animales , Complemento C3/efectos adversos , Factor H de Complemento/administración & dosificación , Terapia Genética , Vectores Genéticos , Humanos , Degeneración Macular/terapia , Ratones , Retina/efectos de los fármacos , Retina/patología
7.
J Gene Med ; 17(6-7): 101-15, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25917932

RESUMEN

BACKGROUND: A variety of disorders are associated with the activation of complement. CD46, CD55 and CD59 are the major membrane associated regulators of complement on human cells. Previously, we have found that independent expression of CD55, CD46 or CD59 through gene transfer protects murine tissues against human complement mediated attack. In the present study, we investigated the potential of combining the complement regulatory properties of CD46, CD55 and CD59 into single gene products expressed from an adeno-associated virus (AAV) vector in a soluble non-membrane anchored form. METHODS: Minigenes encoding the complement regulatory domains from CD46, CD55 and CD59 (SACT) or CD55 and CD59 (DTAC) were cloned into an AAV vector. The specific regulatory activity of each component of SACT and DTAC was measured in vitro. The recombinant AAV vectors were injected into the peritoneum of mice and the efficacy of the transgene products for being able to protect murine liver vasculature against human complement, specifically the membrane attack complex (MAC), was measured. RESULTS: SACT and DTAC exhibited properties similar to CD46, CD55 and CD59 or CD55 and CD59, respectively, in vitro. AAV mediated delivery of SACT or DTAC protected murine liver vasculature from human MAC deposition by 63.2% and 56.7%, respectively. CONCLUSIONS: When delivered to mice in vivo via an AAV vector, SACT and DTAC are capable of limiting human complement mediated damage. SACT and DTAC merit further study as potential therapies for complement mediated disorders when delivered via a gene therapy approach.


Asunto(s)
Antígenos CD55/genética , Antígenos CD59/genética , Activación de Complemento/genética , Inactivadores del Complemento , Proteína Cofactora de Membrana/genética , Transgenes , Animales , Antígenos CD55/química , Antígenos CD59/química , Inactivadores del Complemento/administración & dosificación , Inactivadores del Complemento/química , Dependovirus , Vectores Genéticos , Humanos , Proteína Cofactora de Membrana/química , Ratones , Estructura Terciaria de Proteína/genética
8.
PLoS One ; 8(11): e82295, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278479

RESUMEN

Retinitis pigmentosa (RP) is the most genetically heterogeneous disorder known to cause blindness, involving over 50 different genes. Previously, we have described nanoparticles (NPs) 150 nm in size, comprised of a 3.5 kD peptide (POD) complexed to PEG and DNA (PEGPOD DNA). These NPs expressing GDNF enabled rescue of photoreceptor degeneration in mice up to 11 days post injection. In the current study we examine use of scaffold/ matrix attachment regions (S/MARs), CpG depletion and titration of DNA content of PEGPOD DNA NPs to extend the duration of transgene expression. S/MARs and CpGs did not significantly influence the duration of transgene expression, but did influence its stability. These parameters enabled us to extend transgene expression from 48 hours to 10 weeks. At 77 days post injection, we observed a 76% rescue of the thickness of the retinal outer nuclear layer (ONL) and at 37 days post injection we observed 53% and 55% rescue of the A and B wave ERG amplitudes respectively and 60% rescue of the ONL. Our studies suggest that PEGPOD DNA NPs have potential as gene delivery vectors for the retina.


Asunto(s)
Nanopartículas , Células Fotorreceptoras de Vertebrados/metabolismo , Polietilenglicoles/química , Transgenes , Animales , ADN/metabolismo , Luz , Ratones Endogámicos BALB C , Plásmidos , Reacción en Cadena de la Polimerasa
9.
Invest Ophthalmol Vis Sci ; 54(10): 7107-14, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24106121

RESUMEN

PURPOSE: Immunocytochemical and genetic data implicate a significant role for the activation of complement in the pathology of AMD. Individuals homozygous for a Y402H polymorphism in Factor H have elevated levels of membrane attack complex (MAC) in their choroidal blood vessels and RPE relative to individuals homozygous for the wild-type allele. An R95X polymorphism in C9, a protein necessary for the final assembly of MAC, is partially protective against the formation of choroidal neovascularization (CNV) in AMD patients. Aurintricarboxylic Acid (ATA) is a small molecule inhibitor of MAC. Our hypothesis was that attenuation of the formation of MAC on ocular tissues by ATA may protect mice against laser-induced CNV. METHODS: The ability of ATA to inhibit human complement-mediated cell lysis, inhibit formation of human MAC, and inhibit formation of tubes by endothelial cells was examined in vitro. Subsequently, the Bruch's membrane of adult mice was damaged using an argon laser, followed by intravitreal injection of ATA. One week later, choroidal flat mounts from these mice were stained for the presence of MAC, endothelial cells, and macrophages. RESULTS: ATA protects cells from human complement-mediated lysis, attenuates assembly of the MAC, and inhibits tube formation by endothelial cells in vitro. ATA also attenuates CNV, MAC deposition, and macrophage infiltration in a murine model of exudative AMD. CONCLUSIONS: ATA warrants further study as a potential drug for the treatment of exudative and nonexudative AMD.


Asunto(s)
Ácido Aurintricarboxílico/farmacología , Neovascularización Coroidal/prevención & control , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Degeneración Macular/complicaciones , Adulto , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Humanos , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL
10.
PLoS One ; 8(10): e79661, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24167638

RESUMEN

Diabetic retinopathy is the leading cause of visual dysfunction in working adults and is attributed to retinal vascular and neural cell damage. Recent studies have described elevated levels of membrane attack complex (MAC) and reduced levels of membrane associated complement regulators including CD55 and CD59 in the retina of diabetic retinopathy patients as well as in animal models of this disease. We have previously described the development of a soluble membrane-independent form of CD59 (sCD59) that when delivered via a gene therapy approach using an adeno-associated virus vector (AAV2/8-sCD59) to the eyes of mice, can block MAC deposition and choroidal neovascularization. Here, we examine AAV2/8-sCD59 mediated attenuation of MAC deposition and ensuing complement mediated damage to the retina of mice following streptozotocin (STZ) induced diabetes. We observed a 60% reduction in leakage of retinal blood vessels in diabetic eyes pre-injected with AAV2/8-sCD59 relative to negative control virus injected diabetic eyes. AAV2/8-sCD59 injected eyes also exhibited protection from non-perfusion of retinal blood vessels. In addition, a 200% reduction in retinal ganglion cell apoptosis and a 40% reduction in MAC deposition were documented in diabetic eyes pre-injected with AAV2/8-sCD59 relative to diabetic eyes pre-injected with the control virus. This is the first study characterizing a viral gene therapy intervention that targets MAC in a model of diabetic retinopathy. Use of AAV2/8-sCD59 warrants further exploration as a potential therapy for advanced stages of diabetic retinopathy.


Asunto(s)
Antígenos CD59/biosíntesis , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Dependovirus , Diabetes Mellitus Experimental/terapia , Retinopatía Diabética/terapia , Transducción Genética , Animales , Antígenos CD59/genética , Complejo de Ataque a Membrana del Sistema Complemento/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Humanos , Ratones
11.
PLoS One ; 6(6): e21621, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21720565

RESUMEN

Inappropriate activation of complement on the vascular endothelium of specific organs, or systemically, underlies the etiology of a number of diseases. These disorders include atypical hemolytic uremic syndrome, membranoproliferative glomerulonephritis, atherosclerosis, age-related macular degeneration, diabetic retinopathy, and transplant rejection. Inhibition of the terminal step of complement activation, i.e. formation of the membrane attack complex, using CD59 has the advantage of retaining the upstream processes of the complement cascade necessary for fighting pathogens and retaining complement's crucial role in tissue homeostasis. Previous studies have shown the necessity of membrane targeting of soluble CD59 in order for it to prove an effective inhibitor of complement deposition both in vitro and in vivo. In this study we have generated an in vivo model of human complement activation on murine liver vascular endothelium. This model should prove useful for the development of anti-complement therapies for complement-induced pathologies of vascular endothelium. Using this model, we have demonstrated the viability of a non membrane-targeted soluble CD59 to significantly inhibit complement deposition on the endothelium of murine liver vasculature when expressed in vivo from an adenovirus. This result, unanticipated based on prior studies, suggests that the use of non membrane-targeted sCD59 as an anti-complement therapy be re-visited.


Asunto(s)
Adenoviridae/genética , Antígenos CD59/inmunología , Activación de Complemento/inmunología , Endotelio Vascular/inmunología , Hígado/irrigación sanguínea , Hígado/inmunología , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Aorta/inmunología , Vasos Sanguíneos/citología , Vasos Sanguíneos/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Células Endoteliales/citología , Células Endoteliales/inmunología , Endotelio Vascular/citología , Humanos , Inyecciones Intraperitoneales , Hígado/citología , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Unión Proteica , Suero/inmunología , Solubilidad
12.
PLoS One ; 6(4): e19078, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21552568

RESUMEN

Age related macular degeneration (AMD) is the most common cause of blindness amongst the elderly. Approximately 10% of AMD patients suffer from an advanced form of AMD characterized by choroidal neovascularization (CNV). Recent evidence implicates a significant role for complement in the pathogenesis of AMD. Activation of complement terminates in the incorporation of the membrane attack complex (MAC) in biological membranes and subsequent cell lysis. Elevated levels of MAC have been documented on choroidal blood vessels and retinal pigment epithelium (RPE) of AMD patients. CD59 is a naturally occurring membrane bound inhibitor of MAC formation. Previously we have shown that membrane bound human CD59 delivered to the RPE cells of mice via an adenovirus vector can protect those cells from human complement mediated lysis ex vivo. However, application of those observations to choroidal blood vessels are limited because protection from MAC- mediated lysis was restricted only to the cells originally transduced by the vector. Here we demonstrate that subretinal delivery of an adenovirus vector expressing a transgene for a soluble non-membrane binding form of human CD59 can attenuate the formation of laser-induced choroidal neovascularization and murine MAC formation in mice even when the region of vector delivery is distal to the site of laser induced CNV. Furthermore, this same recombinant transgene delivered to the intravitreal space of mice by an adeno-associated virus vector (AAV) can also attenuate laser-induced CNV. To our knowledge, this is the first demonstration of a non-membrane targeting CD59 having biological potency in any animal model of disease in vivo. We propose that the above approaches warrant further exploration as potential approaches for alleviating complement mediated damage to ocular tissues in AMD.


Asunto(s)
Antígenos CD59/química , Antígenos CD59/genética , Neovascularización Coroidal/genética , Neovascularización Coroidal/terapia , Terapia Genética/métodos , Degeneración Macular/fisiopatología , Degeneración Macular/terapia , Adenoviridae/genética , Animales , Línea Celular , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Humanos , Rayos Láser/efectos adversos , Degeneración Macular/etiología , Degeneración Macular/genética , Ratones , Retina/metabolismo , Solubilidad
13.
Invest Ophthalmol Vis Sci ; 52(6): 3436-45, 2011 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-21357400

RESUMEN

PURPOSE: Activation of complement has been implicated as one of the major causes of age-related macular degeneration (AMD). Evidence is accumulating for a role of complement in other retinal diseases, such as diabetic retinopathy and proliferative vitreoretinopathy. Because of the paucity of animal models that directly investigate the role of complement in retinal pathology, the authors sought to develop a model of increased complement expression and activation, specifically in the murine retina. METHODS: The authors constructed a recombinant adenovirus-expressing murine complement component 3 (C3, AdcmvC3). Adult mice were injected in the subretinal space with either AdcmvC3 or a control virus, AdcmvGFP. After 1 to 2 weeks of exogenous C3 expression, mice were analyzed by scotopic electroretinography and fluorescein angiography. Eyes were harvested for histologic, immunohistochemical, and quantitative RT-PCR analyses. RESULTS: Mice injected with C3-expressing adenovirus exhibited significantly increased vascular permeability, endothelial cell proliferation and migration, RPE atrophy, loss of photoreceptor outer segments, reactive gliosis, retinal detachment, and reduced retinal function relative to those injected with a control adenovirus. Deposition of the membrane attack complex was observed on endothelial cells and photoreceptor outer segments. CONCLUSIONS: Adenovirus-mediated delivery of C3 to murine RPE induces significant functional and anatomic changes that reproduce many of the features of AMD as well as those of other retinal diseases. This novel model may be useful in assessing the role of complement in retinal pathology and in developing anti-complement therapies for retinal diseases associated with complement activation.


Asunto(s)
Adenoviridae/genética , Complemento C3/genética , Regulación de la Expresión Génica/fisiología , Degeneración Macular/genética , Retina/metabolismo , Desprendimiento de Retina/genética , Animales , Atrofia , Permeabilidad Capilar , Movimiento Celular , Proliferación Celular , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Electrorretinografía , Endotelio Vascular/patología , Angiografía con Fluoresceína , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Retina/patología , Desprendimiento de Retina/metabolismo , Desprendimiento de Retina/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodopsina/metabolismo
14.
J Gene Med ; 13(3): 158-70, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21344542

RESUMEN

BACKGROUND: Cell-penetrating peptides (CPPs) can deliver molecules into cells by binding and penetrating the plasma membrane. However, the majority of CPPs get trapped in endosomes, resulting in degradation of the cargo molecule and inefficient delivery to the nucleus. The present study investigates the potential use of a nucleolin binding peptide (NBP) for the delivery of macromolecules including fluorophores, recombinant protein and DNA to the nuclei of ocular tissues in vivo. METHODS: Fluorescent dyes covalently linked to NBP or NBP-green fluorescent protein fusion protein were injected intravitreally or subretinally or topically applied to the cornea. Frozen sections were prepared for quantification of transduction. Delivery of plasmid DNA was studied using luciferase and LacZ DNA compacted with pegylated NBP. Levels of luciferase were quantified, and LacZ expression was localized in ocular tissues. RESULTS: We found that NBP-directed fluorophores exhibited retinal and corneal transduction. Subretinal injection transduced cell types throughout the retina, including photoreceptors, retinal pigment epithelium and neuronal cells. Intravitreal injection transduced neuronal cells in the retina, as well as cells in the cornea. Topically applied NBP lead to transduction of the superficial epithelial layer of the cornea. NBP localized to the nucleus upon exogenous application in vivo. Pegylated NBP nanoparticles significantly improved delivery and expression of transgenes over DNA alone without any measureable toxicity. CONCLUSIONS: The results obtained in the present study demonstrate that NBP can deliver small and large molecules into retinal and corneal cells and plasmid DNA into retinal cells and hence may be useful for the delivery of therapeutics to the eye.


Asunto(s)
Péptidos de Penetración Celular , Córnea/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Proteína HMGN2/administración & dosificación , Péptidos/metabolismo , Polietilenglicoles/metabolismo , Retina/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Administración Tópica , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , Péptidos de Penetración Celular/administración & dosificación , Péptidos de Penetración Celular/metabolismo , ADN/administración & dosificación , ADN/genética , Electrorretinografía , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes , Proteína HMGN2/metabolismo , Humanos , Inyecciones Intravítreas , Luciferasas/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Nanopartículas , Señales de Localización Nuclear/metabolismo , Péptidos/administración & dosificación , Péptidos/genética , Fosfoproteínas/metabolismo , Plásmidos/administración & dosificación , Plásmidos/metabolismo , Polietilenglicoles/administración & dosificación , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , beta-Galactosidasa/genética , Nucleolina
15.
Invest Ophthalmol Vis Sci ; 51(12): 6776-83, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20688727

RESUMEN

PURPOSE: Sequence variations in complement proteins are associated with age-related macular degeneration (AMD). The terminal pathway of complement results in the formation of the membrane attack complex (MAC) on the cell surface, resulting in their lysis. MAC has been documented on the retinal pigment epithelium (RPE), choroidal blood vessels, and drusen of AMD eyes. Here the investigators test the hypothesis that increasing the expression of decay accelerating factor (CD55) on RPE cells may result in reduced MAC-mediated damage. METHODS: The investigators constructed a recombinant adenovirus expressing human CD55 (AdCAGCD55). Mouse hepatocytes were infected with AdCAGCD55 or negative controls and subsequently incubated with normal human serum (NHS). Cell lysis and MAC formation were measured by FACS and immunocytochemistry, respectively. Adult mice were injected in the subretinal space with either AdCAGCD55 or controls; after 1 week of CD55 transgene expression, the eyecups were excised, challenged with NHS, and quantified for human MAC formation. RESULTS: Control-infected or uninfected mouse hepatocytes lyse at a rate of 93% and 94%, respectively. AdCAGCD55- infected mouse hepatocytes lyse at a rate of 29%. Lysis was confirmed to occur in the presence of MAC, which was reduced by 67% when cells were infected by AdCAGCD55. Mice injected in the subretinal space with AdCAGCD55 exhibited a 55.7% reduction in MAC formation on the RPE relative to controls. CONCLUSIONS: Adenovirus-mediated delivery of hCD55 to murine RPE confers protection against human complement. The investigators propose that the expression of hCD55 on RPE cells warrants investigation as a potential therapy for AMD.


Asunto(s)
Antígenos CD55/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/fisiología , Terapia Genética , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/metabolismo , Adenoviridae/genética , Animales , Western Blotting , Antígenos CD55/genética , Línea Celular , Citometría de Flujo , Expresión Génica/fisiología , Vectores Genéticos , Hemólisis/fisiología , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Inmunohistoquímica , Degeneración Macular/metabolismo , Ratones , Ratones Endogámicos C57BL , Transgenes
16.
Mol Ther ; 18(11): 1917-26, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20700110

RESUMEN

Peptide for ocular delivery (POD) is a novel cationic cell-penetrating peptide (CPP) which, when conjugated with polyethylene glycol (PEG-POD), can deliver plasmid DNA to the retinal pigment epithelium (RPE) of adult murine retina. PEG-POD nanoparticles containing an expression cassette for glial cell line-derived neurotrophic factor (PEG-POD~GDNF) were investigated for their ability to inhibit light-induced photoreceptor apoptosis. PEG-POD~GDNF, control nanoparticles, or buffer were injected into the subretinal space of adult murine retina and retinal degeneration induced by blue light. Animals injected with PEG-POD~GDNF showed a significant reduction (3.9-7.7 fold) in apoptosis relative to control-injected animals. The thickness of the outer nuclear layer (ONL) of the superior retina of PEG-POD~GDNF-injected eyes was significantly greater (23.6-39.3%) than control-injected retina 14 days post-light treatment. PEG-POD~GDNF-injected eyes showed a 27-39% greater functional response relative to controls, as measured by electroretinogram (ERG) 7 days post-light treatment. This is one of only two studies demonstrating histological and functional rescue of a mouse model of retinal degeneration following nonviral administration of a transgene into adult retina. Although rescue is short lived for clinical application, this study represents an important step in the development of nonviral gene therapy for retinal diseases.


Asunto(s)
Terapia Genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Nanopartículas , Traumatismos Experimentales por Radiación/terapia , Retina/efectos de la radiación , Degeneración Retiniana/terapia , Animales , Apoptosis , Caspasas/metabolismo , Sistemas de Liberación de Medicamentos , Electrorretinografía , Luz/efectos adversos , Ratones , Ratones Endogámicos BALB C , Fragmentos de Péptidos/uso terapéutico , Polietilenglicoles , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/patología , Ratas , Degeneración Retiniana/etiología , Degeneración Retiniana/patología
17.
Invest Ophthalmol Vis Sci ; 51(12): 6374-80, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20671268

RESUMEN

PURPOSE: Novel zinc finger nucleases (ZFNs) were designed to target the human rhodopsin gene and induce homologous recombination of a donor DNA fragment. METHODS: Three-finger zinc finger nucleases were designed based on previously published guidelines. To assay for ZFN specificity, the authors generated human embryonic retinoblast cell lines stably expressing a Pro23His rhodopsin, the most common mutation associated with autosomal dominant retinitis pigmentosa in North America. They report quantification of these rhodopsin-specific ZFNs to induce a targeted double-strand break in the human genome, demonstrate their ability to induce homologous recombination of a donor DNA fragment, and report the quantification of the frequency of ZFN-mediated homologous recombination. RESULTS: Compared with endogenous homologous recombination, the authors observed a 12-fold increase in homologous recombination and an absolute frequency of ZFN-directed homologous recombination as high as 17% in the human rhodopsin gene. CONCLUSIONS: ZFNs are chimeric proteins with significant potential for the treatment of inherited diseases. In this study, the authors report the design of novel ZFNs targeting the human rhodopsin gene. These ZFNs may be useful for the treatment of retinal diseases such as retinitis pigmentosa, one of the most common causes of inherited blindness in the developed world. Herein, they also report on several aspects of donor fragment design and in vitro conditions that facilitate ZFN-mediated homologous recombination.


Asunto(s)
Endodesoxirribonucleasas/genética , Proteínas Recombinantes de Fusión/genética , Rodopsina/genética , Dedos de Zinc/genética , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica , Ingeniería Genética , Humanos , Mutagénesis Sitio-Dirigida , ARN Mensajero/genética , Recombinación Genética , Retina/embriología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
18.
J Gene Med ; 12(1): 86-96, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19937991

RESUMEN

BACKGROUND: We have previously shown that a novel synthetic peptide for ocular delivery (POD) can efficiently compact DNA and deliver it to cells in vitro. This observation prompted us to develop use of POD as a nonviral vector in vivo. METHODS: POD peptide was modified using poly(ethylene) glycol (PEG-POD) and used to compact DNA into nanoparticles that were then analysed using electron microscopy, dynamic light scattering, and fluorescent labeling. Transfection efficiency and localization were determined 48 h post-injection into the subretinal space of the mouse eye using luciferase and LacZ, respectively. Efficiency of ocular transfection was compared to two other PEGylated peptides: PEG-TAT and PEG-CK30. RESULTS: PEG-POD can compact DNA and form discrete nanoparticles of approximately 136 nm that can penetrate and transduce the retinal pigment epithelium (RPE) in vivo. PEG-POD significantly increased expression of plasmid DNA by 215-fold, PEG-TAT by 56.52-fold, and PEG-CK30 by 24.73-fold relative to DNA injected alone. In all cases beta-galactosidase was observed primarily in the RPE layer after subretinal injection. Electrophysiological analyses of PEG-POD transduced retina indicates an absence of PEG-POD-mediated toxicity. PEG-POD can protect plasmid DNA from DNaseI digestion, resulting in significant transfection of the lung after intravenous injection in mice. CONCLUSIONS: PEG-POD was found to significantly increase gene delivery relative to both DNA alone and other pegylated peptides. These findings highlight the use of pegylated peptides, and specifically PEG-POD, as novel gene delivery vectors.


Asunto(s)
ADN/química , Técnicas de Transferencia de Gen , Mitosis , Nanopartículas/química , Péptidos/química , Polietilenglicoles/química , Epitelio Pigmentado de la Retina/citología , Animales , Línea Celular , ADN/ultraestructura , Desoxirribonucleasa I/metabolismo , Humanos , Inyecciones Intravenosas , Luciferasas/metabolismo , Ratones , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Péptidos/toxicidad , Polietilenglicoles/toxicidad , Estructura Cuaternaria de Proteína , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Transfección
19.
Vision Res ; 50(7): 686-97, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19733192

RESUMEN

Recently we described a novel cell penetrating peptide, peptide for ocular delivery (POD) that could deliver small molecules including fluorescent dyes into retinal cells. The objective of the current study was to examine whether biologically relevant macromolecules such as proteins, genetically fused with POD could also be delivered into retinal tissues in vivo. We generated a POD-GFP fusion protein and examined its cell and tissue penetrating properties. We found that endogenously expressed POD-GFP fusion protein localized to the nucleus, suggesting that POD acts as a nuclear localization signal. Adenovirus (Ad) vectors expressing POD-GFP fusion protein were constructed and the recombinant protein was purified from Ad-infected human embryonic retinoblasts (HER). Exogenously supplied POD-GFP fusion protein rapidly transduced A549 and HER cells and colocalized in part with markers of late endosomes, from which it could escape. Following subretinal delivery, POD-GFP localized to the retinal pigment epithelium and the photoreceptor cell bodies. When injected into the vitreous, POD-GFP localized to the ganglion cells and the inner nuclear layer of the retina as well as the lens capsule. Topical application of POD-GFP to ocular surfaces resulted in uptake by the corneal epithelium. POD-GFP also transduced non-ocular tissues, including the epidermis of the skin following topical application.


Asunto(s)
Córnea/metabolismo , Péptidos/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/metabolismo , Retina/metabolismo , Piel/metabolismo , Adenovirus Humanos/fisiología , Administración Tópica , Transporte Biológico , Humanos , Péptidos/metabolismo , Transfección
20.
Invest Ophthalmol Vis Sci ; 51(4): 2219-28, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19892875

RESUMEN

Purpose. Gene therapy for a number of retinal diseases necessitates efficient transduction of photoreceptor cells. Whereas adenovirus (Ad) serotype 5 (Ad5) does not transduce photoreceptors efficiently, previous studies have demonstrated improved photoreceptor transduction by Ad5 pseudotyped with Ad35 (Ad5/F35) or Ad37 (Ad5/F37) fiber or by the deletion of the RGD domain in the Ad5 penton base (Ad5DeltaRGD). However, each of these constructs contained a different transgene cassette, preventing the evaluation of the relative performance of these vectors, an important consideration before the use of these vectors in the clinic. The aim of this study was to evaluate these vectors in the retina and to attempt photoreceptor-specific transgene expression. Methods. Three Ad5-based vectors containing the same expression cassette were generated and injected into the subretinal space of adult mice. Eyes were analyzed for green fluorescence protein expression in flat-mounts, cross-sections, quantitative RT-PCR, and a modified stereological technique. A 257-bp fragment derived from the mouse opsin promoter was analyzed in the context of photoreceptor-specific transgene expression. Results. Each virus tested efficiently transduced the retinal pigment epithelium. The authors found no evidence that Ad5/F35 or Ad5/F37 transduced photoreceptors. Instead, they found that Ad5/F37 transduced Müller cells. Robust photoreceptor transduction by Ad5DeltaRGD was detected. Photoreceptor-specific transgene expression from the 257-bp mouse opsin promoter in the context of Ad5DeltaRGD vectors was found. Conclusions. Adenovirus vectors may be designed with tropism to distinct cell populations. Robust photoreceptor-specific transgene expression can be achieved in the context of Ad5DeltaRGD vectors.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Neuronas Retinianas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transfección , Animales , Biomarcadores/metabolismo , Western Blotting , Línea Celular Tumoral , Proteínas del Ojo/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica , Proteína Cofactora de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Retina/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA