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

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Hum Gene Ther Clin Dev ; 24(1): 23-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23692380

RESUMEN

Abstract Proof of concept for MERTK gene replacement therapy has been demonstrated using different viral vectors in the Royal College of Surgeon (RCS) rat, a well characterized model of recessive retinitis pigmentosa that contains a mutation in the Mertk gene. MERTK plays a key role in renewal of photoreceptor outer segments (OS) by phagocytosis of shed OS tips. Mutations in MERTK cause impaired phagocytic activity and accumulation of OS debris in the interphotoreceptor space that ultimately leads to photoreceptor cell death. In the present study, we conducted a series of preclinical potency and GLP-compliant safety evaluations of an adeno-associated virus type 2 (AAV2) vector expressing human MERTK cDNA driven by the retinal pigment epithelium-specific, VMD2 promoter. We demonstrate the potency of the vector in RCS rats by improved electroretinogram (ERG) responses in treated eyes compared with contralateral untreated controls. Toxicology and biodistribution studies were performed in Sprague-Dawley (SD) rats injected with two different doses of AAV vectors and buffer control. Delivery of vector in SD rats did not result in a change in ERG amplitudes of rod and cone responses relative to balanced salt solution control-injected eyes, indicating that administration of AAV vector did not adversely affect normal retinal function. In vivo fundoscopic analysis and postmortem retinal morphology of the vector-injected eyes were normal compared with controls. Evaluation of blood smears showed the lack of transformed cells in the treated eyes. All injected eyes and day 1 blood samples were positive for vector genomes, and all peripheral tissues were negative. Our results demonstrate the potency and safety of the AAV2-VMD2-hMERTK vector in animal models tested. A GMP vector has been manufactured and is presently in clinical trial.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Retinitis Pigmentosa/terapia , Animales , Bestrofinas , Canales de Cloruro/genética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Proteínas del Ojo/genética , Femenino , Terapia Genética , Vectores Genéticos/genética , Humanos , Masculino , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/genética , Retina/patología , Retinitis Pigmentosa/patología , Distribución Tisular , Tirosina Quinasa c-Mer
2.
Mol Ther ; 21(3): 509-19, 2013 03.
Artículo en Inglés | MEDLINE | ID: mdl-23358189

RESUMEN

Blindness, although not life threatening, is a debilitating disorder for which few, if any treatments exist. Ocular gene therapies have the potential to profoundly improve the quality of life in patients with inherited retinal disease. As such, tremendous focus has been given to develop such therapies. Several factors make the eye an ideal organ for gene-replacement therapy including its accessibility, immune privilege, small size, compartmentalization, and the existence of a contralateral control. This review will provide a comprehensive summary of (i) existing gene therapy clinical trials for several genetic forms of blindness and (ii) preclinical efficacy and safety studies in a variety of animal models of retinal disease which demonstrate strong potential for clinical application. To be as comprehensive as possible, we include additional proof of concept studies using gene replacement, neurotrophic/neuroprotective, optogenetic, antiangiogenic, or antioxidative stress strategies as well as a description of the current challenges and future directions in the ocular gene therapy field to this review as a supplement.


Asunto(s)
Terapia Genética/métodos , Retina/patología , Enfermedades de la Retina/genética , Enfermedades de la Retina/terapia , Transportadoras de Casetes de Unión a ATP/genética , Animales , Ensayos Clínicos como Asunto , Dependovirus , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Vectores Genéticos , Guanilato Ciclasa/genética , Humanos , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Superficie Celular/genética , Tirosina Quinasa c-Mer , cis-trans-Isomerasas/genética
3.
Mol Vis ; 18: 1668-83, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22773905

RESUMEN

PURPOSE: Although mutated G11778A NADH ubiquinone oxidoreductase subunit 4 (ND4) mitochondrial DNA (mtDNA) is firmly linked to the blindness of Leber hereditary optic neuropathy (LHON), a bona fide animal model system with mutated mtDNA complex I subunits that would enable probing the pathogenesis of optic neuropathy and testing potential avenues for therapy has yet to be developed. METHODS: The mutant human ND4 gene with a guanine to adenine transition at position 11778 with an attached FLAG epitope under control of the mitochondrial heavy strand promoter (HSP) was inserted into a modified self-complementary (sc) adeno-associated virus (AAV) backbone. The HSP-ND4FLAG was directed toward the mitochondria by adding the 23 amino acid cytochrome oxidase subunit 8 (COX8) presequence fused in frame to the N-terminus of green fluorescent protein (GFP) into the AAV2 capsid open reading frame. The packaged scAAV-HSP mutant ND4 was injected into the vitreous cavity of normal mice (OD). Contralateral eyes received scAAV-GFP (OS). Translocation and integration of mutant human ND4 in mouse mitochondria were assessed with PCR, reverse transcription-polymerase chain reaction (RT-PCR), sequencing, immunoblotting, and immunohistochemistry. Visual function was monitored with serial pattern electroretinography (PERG) and in vivo structure with spectral domain optical coherence tomography (OCT). Animals were euthanized at 1 year and processed for light and transmission electron microscopy. RESULTS: The PCR products of the mitochondrial and nuclear DNA extracted from infected retinas and optic nerves gave the expected 500 base pair bands. RT-PCR confirmed transcription of the mutant human ND4 DNA in mice. DNA sequencing confirmed that the PCR and RT-PCR products were mutant human ND4 (OD only). Immunoblotting revealed the expression of mutant ND4FLAG (OD only). Pattern electroretinograms showed a significant decrement in retinal ganglion cell function OD relative to OS at 1 month and 6 months after AAV injections. Spectral domain optical coherence tomography showed optic disc edema starting at 1 month post injection followed by optic nerve head atrophy with marked thinning of the inner retina at 1 year. Histopathology of optic nerve cross sections revealed reductions in the optic nerve diameters of OD versus OS where transmission electron microscopy revealed significant loss of optic nerve axons in mutant ND4 injected eyes where some remaining axons were still in various stages of irreversible degeneration with electron dense aggregation. Electron lucent mitochondria accumulated in swollen axons where fusion of mitochondria was also evident. CONCLUSIONS: Due to the UGA codon at amino acid 16, mutant G11778A ND4 was translated only in the mitochondria where its expression led to significant loss of visual function, loss of retinal ganglion cells, and optic nerve degeneration recapitulating the hallmarks of human LHON.


Asunto(s)
Ceguera/genética , Dependovirus/genética , Mitocondrias/genética , NADH Deshidrogenasa/genética , Atrofia Óptica Hereditaria de Leber/genética , Atrofia Óptica/genética , Animales , Ceguera/enzimología , Ceguera/patología , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/genética , Electrorretinografía , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Inyecciones Intravítreas , Ratones , Mitocondrias/enzimología , NADH Deshidrogenasa/metabolismo , Atrofia Óptica/enzimología , Atrofia Óptica/patología , Atrofia Óptica Hereditaria de Leber/enzimología , Atrofia Óptica Hereditaria de Leber/patología , Nervio Óptico/enzimología , Nervio Óptico/patología , Mutación Puntual , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Ganglionares de la Retina/enzimología , Células Ganglionares de la Retina/patología
4.
Hum Mol Genet ; 20(23): 4569-81, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21880665

RESUMEN

Defects in the photoreceptor-specific gene aryl hydrocarbon receptor interacting protein-like 1 (Aipl1) are associated with Leber congenital amaurosis (LCA), a childhood blinding disease with early-onset retinal degeneration and vision loss. Furthermore, Aipl1 defects are characterized at the most severe end of the LCA spectrum. The rapid photoreceptor degeneration and vision loss observed in the LCA patient population are mimicked in a mouse model lacking AIPL1. Using this model, we evaluated if gene replacement therapy using recent advancements in adeno-associated viral vectors (AAV) provides advantages in preventing rapid retinal degeneration. Specifically, we demonstrated that the novel self-complementary Y733F capsid mutant AAV2/8 (sc-Y733F-AAV) provided greater preservation of photoreceptors and functional vision in Aipl1 null mice compared with single-stranded AAV2/8. The benefits of sc-Y733F-AAV were evident following viral administration during the active phase of retinal degeneration, where only sc-Y733F-AAV treatment achieved functional vision rescue. This result was likely due to higher and earlier onset of Aipl1 expression. Based on our studies, we conclude that the sc-Y733F-AAV2/8 viral vector, to date, achieves the best rescue for rapid retinal degeneration in Aipl1 null mice. Our results provide important considerations for viral vectors to be used in future gene therapy clinical trials targeting a wider severity spectrum of inherited retinal dystrophies.


Asunto(s)
Cápside/metabolismo , Dependovirus/genética , Terapia Genética , Amaurosis Congénita de Leber/fisiopatología , Amaurosis Congénita de Leber/terapia , Mutación/genética , Visión Ocular/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Conducta Animal , Proteínas de la Cápside/genética , Proteínas Portadoras/genética , Proteínas Portadoras/uso terapéutico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Dependovirus/efectos de la radiación , Proteínas del Ojo/genética , Proteínas del Ojo/uso terapéutico , Humanos , Amaurosis Congénita de Leber/complicaciones , Amaurosis Congénita de Leber/patología , Luz , Ratones , Retina/enzimología , Retina/patología , Retina/efectos de la radiación , Degeneración Retiniana/complicaciones , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Degeneración Retiniana/terapia , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Visión Ocular/efectos de la radiación
5.
Mol Vis ; 17: 1090-102, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21552473

RESUMEN

PURPOSE: With the increasing number of retinal gene-based therapies and therapeutic constructs, in vitro bioassays characterizing vector transduction efficiency and quality are becoming increasingly important. Currently, in vitro assays quantifying vector transduction efficiency are performed predominantly for non-ocular tissues. A human retinal pigment epithelial cell line (ARPE19) and a mouse cone photoreceptor cell line, 661W, have been well characterized and are used for many retinal metabolism and biologic pathway studies. The purpose of this study is to quantify transduction efficiencies of a variety of self-complementary (sc) adeno-associated virus (AAV) vectors in these biologically relevant ocular cell lines using high-throughput fluorescence-activated cell sorting (FACS) analysis. METHODS: ARPE19 and 661W cells were infected with sc-smCBA-mCherry packaged in unmodified AAV capsids or capsids containing single/multiple tyrosine-phenylalanine (Y-F) mutations at multiplicity of infections (MOIs) ranging from 100 to 10,000. Three days post infection fluorescent images verified mCherry expression. Following microscopy, FACS analysis was performed to quantify the number of positive cells and the mean intensity of mCherry fluorescence, the product of which is reported as transduction efficiency for each vector. The scAAV vectors containing cone-specific (sc-mCARpro-green fluorescent protein [GFP]), rod-specific (sc-MOPS500-eGFP), retinal pigment epithelium (RPE)-specific (sc-VMD2-GFP), or ubiquitous (sc-smCBA-GFP) promoters were used to infect both cell lines at an MOI of 10,000. Three days post infection, cells were immunostained with an antibody raised against GFP and imaged. Finally, based on our in vitro results, we tested a prediction of transduction efficiency in vivo. RESULTS: Expression from unmodified scAAV1, scAAV2, scAAV5, and scAAV8 vectors was detectable by FACS in both ARPE19 and 661W cells, with scAAV1 and scAAV2 being the most efficient in both cell lines. scAAV5 showed moderate efficiency in both ARPE19 and 661W cells. scAAV8 was moderately efficient in 661W cells and was by comparison less so in ARPE19 cells; however, transduction was still apparent. scAAV9 performed poorly in both cell types. With some exceptions, the Y-F capsid mutations generally increased the efficiency of scAAV vector transduction, with the increasing number of mutated residues improving efficiency. Results for single scAAV1 and scAAV8 capsid mutants were mixed. In some cases, efficiency improved; in others, it was unchanged or marginally reduced. Retinal-specific promoters were also active in both cell lines, with the 661W cells showing a pattern consistent with the in vivo activity of the respective promoters tested. The prediction based on in vitro data that AAV2 sextuple Y-F mutants would show higher transduction efficiency in RPE relative to AAV2 triple Y-F capsid mutants was validated by evaluating the transduction characteristics of the two mutant vectors in mouse retina. CONCLUSIONS: Our results suggest that this rapid and quantifiable cell-based assay using two biologically relevant ocular cell lines will prove useful in screening and optimizing AAV vectors for application in retina-targeted gene therapies.


Asunto(s)
Dependovirus/metabolismo , Células Epiteliales/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células Fotorreceptoras/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Retina/metabolismo , Animales , Cápside/química , Cápside/metabolismo , Línea Celular , Dependovirus/genética , Células Epiteliales/citología , Citometría de Flujo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/química , Proteínas Fluorescentes Verdes/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Fenilalanina/genética , Fenilalanina/metabolismo , Células Fotorreceptoras/citología , Proteínas Recombinantes de Fusión/genética , Retina/citología , Transformación Genética , Transgenes , Tirosina/genética , Tirosina/metabolismo , Visión Ocular
6.
Arch Ophthalmol ; 128(7): 876-83, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20625049

RESUMEN

OBJECTIVE: To demonstrate the high efficiency and rapidity of allotopic expression of a normal human ND4 subunit of complex I in the vertebrate retina using a self-complementary adeno-associated virus (scAAV) vector for ocular gene delivery to treat acute visual loss in Leber hereditary optic neuropathy (LHON). METHODS: The nuclear-encoded human ND4 subunit fused to the P1 isoform of subunit C of adenosine triphosphate synthase (ATPc) mitochondrial targeting sequence and FLAG epitope was packaged in scAAV2 capsids or single-stranded (ss) AAV2 capsids. These constructs were injected into the vitreous cavities of mice. The contralateral eyes were injected with scAAV-green fluorescent protein (GFP). One week later, pattern electroretinograms and gene expression of the human ND4 subunit and GFP were evaluated. Quantitative analysis of ND4FLAG-injected eyes was assessed relative to Thy1.2-labeled retinal ganglion cells (RGCs). RESULTS: Pattern electroretinogram amplitudes remained normal in eyes inoculated with scAAV-ND4FLAG, ssAAV-ND4FLAG, and GFP. Confocal microscopy revealed the typical perinuclear mitochondrial expression of scAAV-ND4FLAG in almost the entire retinal flat mount. In contrast, scAAV-GFP expression was cytoplasmic and nuclear. Relative to Thy1.2-positive RGCs, quantification of scAAV-ND4FLAG-positive RGCs was 91% and that of ssAAV-ND4FLAG-positive RGCs was 51%. CONCLUSION: Treatment of acute visual loss due to LHON may be possible with a normal human ND4 subunit gene of complex I, mutated in most cases of LHON, when delivered by an scAAV vector. Clinical Relevance Unlike most retinal degenerations that result in slowly progressive loss of vision over many years, LHON due to mutated mitochondrial DNA results in apoplectic, bilateral severe and usually irreversible visual loss. For rescue of acute visual loss in LHON, a highly efficient and rapid gene expression system is required.


Asunto(s)
Dependovirus/genética , Complejo I de Transporte de Electrón/genética , Regulación de la Expresión Génica/fisiología , Vectores Genéticos , NADH Deshidrogenasa/genética , Células Ganglionares de la Retina/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Complejo I de Transporte de Electrón/metabolismo , Electrorretinografía , Técnica del Anticuerpo Fluorescente Indirecta , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos DBA , Microscopía Confocal , NADH Deshidrogenasa/metabolismo , Antígenos Thy-1/genética , Agudeza Visual
7.
Mol Ther ; 12(6): 1072-82, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16226919

RESUMEN

The short- and long-term effects of gene therapy using AAV-mediated RPE65 transfer to canine retinal pigment epithelium were investigated in dogs affected with disease caused by RPE65 deficiency. Results with AAV 2/2, 2/1, and 2/5 vector pseudotypes, human or canine RPE65 cDNA, and constitutive or tissue-specific promoters were similar. Subretinally administered vectors restored retinal function in 23 of 26 eyes, but intravitreal injections consistently did not. Photoreceptoral and postreceptoral function in both rod and cone systems improved with therapy. In dogs followed electroretinographically for 3 years, responses remained stable. Biochemical analysis of retinal retinoids indicates that mutant dogs have no detectable 11-cis-retinal, but markedly elevated retinyl esters. Subretinal AAV-RPE65 treatment resulted in detectable 11-cis-retinal expression, limited to treated areas. RPE65 protein expression was limited to retinal pigment epithelium of treated areas. Subretinal AAV-RPE65 vector is well tolerated and does not elicit high antibody levels to the vector or the protein in ocular fluids or serum. In long-term studies, wild-type cDNA is expressed only in target cells. Successful, stable restoration of rod and cone photoreceptor function in these dogs has important implications for treatment of human patients affected with Leber congenital amaurosis caused by RPE65 mutations.


Asunto(s)
Ceguera/genética , Ceguera/terapia , Dependovirus/genética , Terapia Genética/métodos , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Animales , Animales Modificados Genéticamente , Western Blotting , Proteínas Portadoras , Cromatografía , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Perros , Electrorretinografía , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo/genética , Eliminación de Gen , Técnicas de Transferencia de Gen , Vectores Genéticos , Homocigoto , Humanos , Inmunohistoquímica , Mutación , Regiones Promotoras Genéticas , Degeneración Retiniana/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transgenes , cis-trans-Isomerasas
8.
PLoS Med ; 2(11): e333, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16250670

RESUMEN

BACKGROUND: Leber congenital amaurosis (LCA), a heterogeneous early-onset retinal dystrophy, accounts for approximately 15% of inherited congenital blindness. One cause of LCA is loss of the enzyme lecithin:retinol acyl transferase (LRAT), which is required for regeneration of the visual photopigment in the retina. METHODS AND FINDINGS: An animal model of LCA, the Lrat-/- mouse, recapitulates clinical features of the human disease. Here, we report that two interventions--intraocular gene therapy and oral pharmacologic treatment with novel retinoid compounds--each restore retinal function to Lrat-/- mice. Gene therapy using intraocular injection of recombinant adeno-associated virus carrying the Lrat gene successfully restored electroretinographic responses to approximately 50% of wild-type levels (p < 0.05 versus wild-type and knockout controls), and pupillary light responses (PLRs) of Lrat-/- mice increased approximately 2.5 log units (p < 0.05). Pharmacological intervention with orally administered pro-drugs 9-cis-retinyl acetate and 9-cis-retinyl succinate (which chemically bypass the LRAT-catalyzed step in chromophore regeneration) also caused long-lasting restoration of retinal function in LRAT-deficient mice and increased ERG response from approximately 5% of wild-type levels in Lrat-/- mice to approximately 50% of wild-type levels in treated Lrat-/- mice (p < 0.05 versus wild-type and knockout controls). The interventions produced markedly increased levels of visual pigment from undetectable levels to 600 pmoles per eye in retinoid treated mice, and approximately 1,000-fold improvements in PLR and electroretinogram sensitivity. The techniques were complementary when combined. CONCLUSION: Intraocular gene therapy and pharmacologic bypass provide highly effective and complementary means for restoring retinal function in this animal model of human hereditary blindness. These complementary methods offer hope of developing treatment to restore vision in humans with certain forms of hereditary congenital blindness.


Asunto(s)
Aciltransferasas/genética , Ceguera/tratamiento farmacológico , Ceguera/genética , Terapia Genética , Atrofia Óptica Hereditaria de Leber/tratamiento farmacológico , Atrofia Óptica Hereditaria de Leber/genética , Profármacos/farmacología , Vitamina A/análogos & derivados , Adenoviridae/genética , Administración Oral , Animales , Modelos Animales de Enfermedad , Diterpenos , Vectores Genéticos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Profármacos/administración & dosificación , Profármacos/uso terapéutico , Pupila/fisiología , Pigmentos Retinianos/análisis , Ésteres de Retinilo , Vitamina A/administración & dosificación , Vitamina A/farmacología , Vitamina A/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA